a delay in consolidation is observed in a heterozygous...

101
Noggin and Chordin knockdown in Distraction Osteogenesis Priscille Grenier-Vallée, MD Department of Experimental Surgery McGill University Montreal, Québec Canada April 2013 A thesis submitted to McGill University, Faculty of Graduate and Postdoctoral Studies, in partial fulfillment of the requirements of the degree of Masters of Science in Experimental Surgery

Upload: tranhanh

Post on 16-Sep-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Noggin and Chordin knockdown in Distraction Osteogenesis

Priscille Grenier-Vallée, MD

Department of Experimental Surgery

McGill University

Montreal, Québec Canada

April 2013

A Delay in Consolidation is Observed in a Heterozygous Conditional BMP2 Deficient Mouse Model of Distraction

Osteogenesis

Norine Alam

Department of Human Genetics McGill University Montreal, Québec

Canada

February 2009

A thesis submitted to McGill University, Faculty of Graduate and Postdoctoral Studies, in partial fulfillment of the requirements of the

degree of Masters of Science in Human Genetics

©Copyright Norine Alam, 2009.

1

A thesis submitted to McGill University, Faculty of Graduate and Postdoctoral Studies, in partial fulfillment of the requirements of the degree of Masters of Science in

Experimental Surgery

ABSTRACT

Introduction: Numerous reports have shown that recombinant BMPs have positive effects in several conditions associated with poor bone formation. We hypothesize that by inhibiting BMP antagonists Noggin and Chordin using RNA interference we may upregulate endogenous BMP expression and enhance osteogenesis.

Methods: MC3T3-E1 cells were transduced with lentiviruses expressing various shRNAs targeting the mouse Noggin (5 shRNAs) and Chordin (3 shRNAs) genes and 1 negative control. At various time points after infection, levels of RNA expression for Noggin and Chordin were monitored through RT-PCR. Western blotting were performed on the cell extracts and culture media to verify the expression and secretion of Noggin and Chordin proteins. Cell extracts were also analyzed on day 2 and 4 after transduction for alkaline phosphastase activity which is a marker of osteogenic differentiation.

DO was performed on the right tibia of 54 wild-type mice using a miniature Illizarov distraction device. Animals were randomized into two major groups according to the time of sacrifice: end of distraction (day 17) and mid-consolidation (day 34). Each major group was sub-divided in 3 subgroups representing the type of injection that was administered at the site of distraction on day 8. For each time point, 9 mice were injected with PBS, 9 with a lentivirus plasmid containing a Non-Target (NT) shRNA and 9 with lentivirus plasmids containing shRNAs targeting Noggin-Chordin. To evaluate the success of the infection a GFP marker was added to lentivirus plasmid targeting Noggin-Chordin. For each subgroup 6 collected samples were studied using Faxitron X-ray, µCT and immunohistochemistry and 3 were studied using Rt-PCR.

Results: On non-tranduced MC3T3-E1 cells, the levels of RNA expression of Noggin and Chordin was at its highest level on Day 7. At this timepoint, qRT-PCR analysis showed that the shRNAs were effective in knocking down Noggin and Chordin endogenous mRNA levels down to 10% and 17%, respectively, by the most potent of shRNAs tested compared to control. Western Blot analysis also corroborates that the respective shRNAs were effective in knocking down the Noggin and Chordin proteins. Specific activity of alkaline phosphatase was increased in MC3T3-E1 cells stably expressing Noggin and Chordin shRNA.

Concerning our in vivo work, all the surgeries and distraction were successful except for the death of 2 animals secondary to post-operative complications. However, at the time of the injection the majority of the injection content was extravasating out of the injection site. Once the samples collected, the Faxitron X-ray and µCT studies did not show a significant difference of bone volume or in the ratio of bone volume/tissue volume at the distraction site in between the 3 injection subgroups at both time points. The Rt-PCR studies did not show a significant difference in inhibiting Noggin or Chordin mRNA levels in between the 3 subgroups at both time points. Finally, the immunohistochemistry studies were stopped after the GFP marker was found in all the three subgroups.

2

Conclusion: In our in vitro study, western blot analysis, qRT-PCR and ALP assay were consistent with a successful knockdown of Noggin and Chordin.

During our in vivo study, the main issues faced during this project were technical problems during the injection. Unfortunately, our study did not show any significant difference in bone formation after using injection of lentivirus plasmid with shRNA targetting BMPs antagonists Noggin and Chordin.

3

RÉSUMÉ

Introduction: Plusieurs études ont demontré que l’utilisation des BMPs ont des effets positifs sur plusieurs conditions associées avec une mauvaise formation osseuse. Notre hypothèse est qu’en inhibant les antagonites des BMPs Noggin et Chordin en utilisant une technique d’interférence de l’ARN nous pourrions augmenter l’expression endogène des BMP et par le fait même augmenter l’osteogénèse.

Methodes: Les cellules MC3T3-E1 furent transfectées avec les lentivirus qui expriment différents shRNAs visant Noggin (5 shRNAs) et Chordin (3 shRNAS) chez la souris. Un contrôle négatif fut aussi sélectionné, NT (Non-Target shRNA). À différents temps après l’infection, les niveau d’expression d’ARN furent mesurés à l’aide d’un RT-PCR. Des études de Western blot furent performées sur les extraits cellulaires et sur les milieux de culture pour vérifier l’expression et la sécrétion des protéines Noggin et Chordin. Des extraits cellulaires furent aussi analysés au jour 2 et 4 après l’infection pour mesurer l’activité de l’alkaline phosphatase, qui est un marqueur de la différentiation osteogenique.

Par la suite, une distraction osseuse a été réalisée sur le tibia droit de 54 souris de type sauvage à l’aide d’un miniature fixateur externe Ilizarov. Les animaux étaient randomisés en 2 groupes selon leur temps de sacrifice : fin de la distraction (Jour 17) et mi-consolidation (Jour 34). Chaque groupe était sous-divisé en 3 sous-groupes représentant le type d’injection qui était administrée au site de distraction au jour 8. Pour chaque temps de sacrifice, 9 souris étaient injectées avec du PBS, 9 souris avec un Non-Target shRNA (NT) et 9 souris avec des lentivirus contenant Noggin et Chordin. Pour évaluer le succès notre infection, le shRNA qui visait Noggin fut transféré dans un plasmide contenant un marqueur GFP. Aussi, pour chaque sous-groupe, 6 échantillons étaient collectés pour une étude au Faxitron X-ray, µCT and immunohistochimie et 3 échantillons furent étudiés à l’aide du RT-PCR.

Resultats: Sur les cultures de MC3T3-E1 non transfectées, les niveaux d’expression d’ARN pour Noggin et Chordin étaient à leur plus haut niveau au Jour 7. Au Jour 7, les études au RT-PCR ont aussi démontré que les lentivirus contenant les shRNAs les plus efficaces avaient créé une diminution des niveaux endogènes d’expression d’ARN de Noggin et Chordin à 10 et 17% respectivement comparés au contrôle. Les analyses de Western Blot ont aussi confirmé que ces mêmes shRNAs avaient réussi une diminution significative des protéines de Noggin et Chordin. Aussi, l’activité de l’alkaline phosphatase était augmentée par les cultures exprimant de façon stable les shRNAs visant Noggin et Chordin.

Pour les études in vivo, toutes les chirurgies et les distraction furent réussies à l’exception de 2 morts suite à des complications chirurgicales. Toutefois, au moment de l’injection la majorité du contenu de l’injection However, at the time of the injection the majority of the injection ressortait du site d’injection. Une fois les échantillons collectés, les études Faxitron X-ray et µCT n’ont pas démontré de différence significative entre les 3 groupes dans le volume osseux ou le ration du volume osseux/volume tissulaire et ce, aux 2 temps de sacrifices. Finalement, les études en

4

immunohistochimie furent arrêtées après qu’un marquage au GFP fut retrouvé dans les 3 groupes.

Conclusion: Les analyses de Western blot analysis, qRT-PCR et les essais d’ ALP assay sont en accord avec un knockdown réussi de Noggin et Chordin lors de notre étude in vitro.

Lors de notre étude in vivo, les principaux problèmes rencontrés furent reliés à la technique de l’injection. Malheureuse, cette partie de notre étude n’a pas pu démontrer de différence significative dans la formation osseuse après l’utilisation d’injection de lentivirus contenant des shRNAs visant les antagonistes des BMPs Noggin et Chordin.

5

ACKNOWLEDGEMENTS

I would like to greatly thank my supervisors, Dr. Reggie Hamdy (Orthopedic Surgery) that took

me under his wing when I needed to take the next important step in my career and Dr Pierre

Moffatt ( Human Genetics ) for his constant support, guidance, and availability.

In addition, I would like to thank Dr. Abdallah Husseini for his help for the surgical procedures.

Thanks to the members of the McGill Bone Center for Micro-CT, Faxitron X-ray, and

biomechanical testing analysis.

My sincere gratitude to Bahar Kasaii with her help in coordinating all the experiments and

analyzing the results.

I would like to also thank the Shriners animal technicians, Mia, and Nathalie for their assistance

with distractions in the animal model.

Lastly, I would like to thank my parents, my brother and my fiance for their constant support and

help without whom I would never be able to reach all of my goals.

6

TABLE OF CONTENTS

ABSTRACT ................................................................................................................................. 2

RÉSUMÉ .......................................................................................................................................4

ACKNOWLEDGEMENTS ......................................................................................................... 6

TABLE OF CONTENTS .............................................................................................................. 7

LIST OF FIGURES ......................................................................................................................11

LIST of GRAPHICS......................................................................................................................13

LIST LIST OF TABLES .............................................................................................................. 14

LIST OF ABBREVIATIONS ...................................................................................................... 15

1-INTRODUCTION .................................................................................................................. 17

1.1-Distraction osteogenesis............................................................................................. 17

1.1.1-History..........................................................................................................19

1.1.2-Principle of tension-stress........................................................................... 19

1.1.3-Phases of distraction osteogenesis...............................................................20

1.1.4-Bone development during DO ................................................................... 22

1.1.5-Types of ossification in DO.........................................................................23

1.6-Complications and drawbacks of DO............................................................ 24

1.2-Attempts to accelerate................................................................................................ 25

1.2.1-Mechanical Stimulation ............................................................................. 26

1.2.2- Low-Intensity Pulse Ultrasound (LIPUS).................................................. 26

1.2.3-Transplantation of Bone Marrow Cells (BMCs)......................................... 28

1.2.4- Transplantation of Osteoblastlike cells ......................................................29

1.2.5- Demineralized Bone Matrix........................................................................31

7

1.2.6- Bone-graft substitutes..................................................................................31

1.2.7-Bisphosphonates ..........................................................................................33

1.2.8-Growth Factors............................................................................................ 34

1.2.8.1 - Growth Hormone Injection..........................................................34

1.2.9 - Cartilage Oligomeric Matrix Protein - angiopoietin-1 (COMP-Ang1)......34

1.2.10- Bone Morphogenetic Proteins(BMPS) .....................................................35

1.2.10.1 - BMPs Function......................................................................... 36

2.10.2- The BMP pathway........................................................................ 37

1.2.10.3 Use of exogenous BMPs during Distraction Osteogenesis..........40

1.2.10.3.1 Complications usage of exogenous BMPs....................41

1.2.11 -BMP antagonists....................................................................................... 42

1.2.11.1 Noggin.........................................................................................42

1.2.11.2 Chordin........................................................................................43

1.2.12 RNA interference (RNAi)...........................................................................44

1.2.12.1 shRNA.........................................................................................46

1.2.12.2 Delivery systems shRNA.............................................................48

1.2.12.2.1 Lentiviral vector............................................................48

2 - OBJECTIVE OF STUDY .................................................................................................... 50

3 -MATERIALS & METHODS IN VITRO STUDIES .......................................................... 51

3.1-shRNA selection .........................................................................................................51

3.2-Lentiviruses production.............................................................................................. 52

3.3- MC3T3-E1 transduction............................................................................................ 53

3.4-RT-PCR study............................................................................................................. 54

3.4.1 -RNA extraction from cell cultures............................................................. 54

8

3.4.2-RNA purification ........................................................................................54

3.4.3-RNA Quantification.................................................................................... 55

3.4.4-Reverse transcription of RNA......................................................................55

3.4.5- Real-Time PCR...........................................................................................55

3.5- Western Blot study.....................................................................................................56

3.5.1 - Proteins collection from cell culture and media........................................56

3.5.2 - Samples preparation..................................................................................57

3.5.3 - Western Blot..............................................................................................57

3.6 - Alkaline phosphatase assay......................................................................................58

4 - MATERIALS & METHODS IN VIVO STUDIES............................................................59

4.1 - Operative and distraction protocol...........................................................................60

4.2 - Injection....................................................................................................................63

4.2.1- Transfer of Noggin shRNA into lentivirus plasmid containing GFP..........63

4.2.2 - Injection protocol.......................................................................................64

4.3 - Faxitron X-ray and Micro-Computed Tomography..................................................64

4.4 - Immunohistochemistry..............................................................................................65

4.4.1- Paraffin embedding.....................................................................................65

4.4.2 - GFP marking..............................................................................................66

4.5- RT-PCR on tibia samples...........................................................................................66

4.5.1- Bone collection...........................................................................................66

4.5.2 - Tissue Homogenisation.............................................................................66

4.5.3 - RNA extraction, reverse transcription RNA and RT-PCR ........................67

4.5.4- RT-PCR : GFP and puromycin...................................................................67

9

5- IN VITRO RESULTS..............................................................................................................68

5.1 - RT-PCR results non-transfected MC3T3-E1.............................................................68

5.2 - RT-PCR transduced MC3T3-E1 cultures..................................................................69

5.3 - Western blot...............................................................................................................71

5.4 - Phosphatase alkaline assay........................................................................................74

6- IN VIVO RESULTS................................................................................................................76

6.1- Surgical and distraction protocol success rate...........................................................76

6.2 - Injection success rate................................................................................................76

6.3 - Faxitron X-Ray results.............................................................................................76

6.4 - Micro-Computed Tomography (µ-CT).....................................................................79

6.4.1 - Bone volume..............................................................................................79

6.4.2 - Bone volume/ Tissue volume ratio.............................................................81

6.5 - RT-PCR tibia samples...............................................................................................82

6.6-Immunohistochemistry - GFP marking.......................................................................83

7- DISCUSSION...........................................................................................................................85

8- REFERENCES ...................................................................................................................... 89

10

LIST OF FIGURES

Figure 1: Ilizarov external fixator on human patient

Figure 2: Phase of distraction osteogenesis.

Figure 3: BMP signaling pathway.

Figure 4: shRNA mediated RNA interference pathway

Figure 5: Position of shRNAs

Figure 7: Lentiviral plasmid

Figure 8: Mini Ilizarov external fixator

Figure 9: Mouse in DO

Figure 10: Final plasmid containing GFP after Noggin shRNA transfer

Figure 11 : Noggin - Western blot on secreted proteins in condition media of MC3T3-E1 stably expressing shRNAs targeting Noggin

Figure 12 : Noggin - Western blot on cellular proteins of MC3T3-E1 stably expressing shRNAs targeting Noggin

11

Figure 13: Chordin - Western blot secreted proteins in condition media of MC3T3-E1 stably expressing shRNAs targeting Chordin

Figure 14 : Puromycin and GFP signal in MC3T3-E1 cells stably expressing shRNAs targeting Noggin and Chordin

12

LIST OF GRAPHICS

Graph 1 : RT-PCR -MC3T3-E1 Noggin and Chordin mRNA level

Graph 2 : Noggin mRNA levels in stable pools MC3T3-E1 expressing individual shRNA through lentiviral infection

Graph 3 : Chordin mRNA levels in stable pools MC3T3-E1 expressing individual shRNA through lentiviral infection

Graph 4 : ALP activity in MC3T3-E1 stably expressing the shRNA targeting Noggin and Chordin

Graph 5 : Bone filling scores

Graph 6 : Bone volumes

Graph 7 : Bone volume / Tissue volume ratios

Graph 8 : RT-PCR tibia samples

13

LIST OF TABLES

Table 1 : Timeline animal procedures during in vivo studies

Table 2 : Bone filling scores

14

LIST OF ABBREVIATIONS

µCT Micro-computed tomography

ActR Activin receptor

ATF-4 Activating transcription factor-4

ALK Activin receptor-like kinase

ALP Alkaline phosphotase

BMC Bone marrow cell

BMP Bone morphogenetic protein

BMPR Bone morphogenetic protein receptor

BSA Bovine serum albumin

BV Bone volume

BV/TV Bone volume/tissue volume ratio

cDNA Complementary deoxyribonucleic acid

co-SMAD Common-partner mothers against decapentaplegic homolog

Col1a1 Collagen type I

DEPC Diethylpyrocarbonate

DO Distraction osteogenesis

DTT Dithiothreitol

EDTA Ethylenediaminetetraacetic acid

FGF Fibroblast growth factor

IGF Insulin-like growth factor

I-SMAD Inhibitory mothers against decapentaplegic homolog

LIPUS Low-intensity pulse ultrasound

mRNA Messenger ribonucleic acid NaCl Sodium chloride

15

PBS Phosphate buffer saline

PCR Polymerase chain reaction

PDGF Platelet-derived growth factor

PFA Paraformaldehyde

PRP Platelet-rich plasma

R-SMAD Receptor-regulated mothers against decapentaplegic homolog

RT-qPCR Real Time quantitative polymerase chain reaction

rhBMP Recombinant human bone morphogenetic protein

RNA Ribonucleic acid

SD Standard deviation

SMAD Mothers against decapentaplegic homolog

SMURF Smad ubiqutin regulatory factor

TBS-T Tri-phosphate buffer saline-tween TCA Trichloroacetic acid

TGF-ß Transforming growth factor-ß

TRIS Hydroxymethylaminomethane

µCT Micro-computed tomography

16

1- INTRODUCTION

1.1-Distraction osteogenesis

Distraction osteogenesis (DO) is a surgical procedure and treatment for lengthening of bone

adressing bony deficiencies secondary to congenital deformities, traumatic bone loss, infection,

malignancy and congenital limb discrepancies. Distraction osteogenesis is performed with a

transverse osteotomy. The fracture bone is then kept in position with an external fixator. During

the process the fractured sites are slowly pulled apart for a specific amount of time. The patients

bone regenerative properties will form new remodeled bone in the distraction gap. [1-2]

17

Figure 1 : Ilizarov external fixator on human patient

Schematic presentation of Ilizarov apparatus for human tibial distraction lengthening.

18

1.1.1-History

Long bone lengthening has been undertaken for more than 100 years, dating back to Codvilla

who performed femoral lengthening using traction after femoral osteotomy, using a pin placed in

calcaneus. [3]

Eventually, bone distraction evolved into different modern techniques. The Illizarov technique is

certainly one of the most well-known. In 1951, Dr. Gavriil Abramovitch Ilizarov developed the

surgical technique of distraction osteogenesis. (Figure 1) Ilizarov first had his idea of DO when a

patient mistakenly started turning the rods of his circular external fixator. Ilizarov noticed that

progressively, new bone had developed within the fractured gap. Intrigued by this biological

finding and its potential use in clinical treatment, Ilizarov began researching the technique using

animal models. In 1980, Ilizarov used DO to treated Carlo Mauri’s, an Italian explorer who was

suffering of pseudoarthrosis Mauri was very impressed with the results of his surgery and he set

out with Ilizarov to introduce this novel method of fracture treatment to the orthopedic

community. A few years after, DO gained public recognition and was commonly practiced. [4]

1.1.2-The Principle of Tension-Stress

The Principle of Tension-Stress was developed by Ilizarov and his team. This principle explains

the bone regeneration during DO. According to them, the slow and steady traction of a tissue

metabolically activates biosynthetic and proliferative pathways involved in bone regeneration

within the distracted zone. Many factors are involved in the bone quality that is developed at the

19

distraction site : the stability of the external fixator, the quality of tissues around the distraction

site and the rate and the frequency of distraction. [2-4]

1.1.3-Phases of Distraction Osteogenesis

Distraction osteogenesis is divided into three major phases: the latency phase, the distraction

phase and the consolidation phase. (Figure 2) The latency phase starts the first day post surgery

and lasts 5 to 7 days. Then the distraction is debuted and usually performed at a rate of 1mm/day

in humans . The act of distraction enhances angiogenic activity, activating intracellular pathways

at the distraction site. As distraction progresses, fibroblast-like cells grow parallel to the direction

of the tension-stress vector in the lengthened gap. The fibroblast-like cells produce collagen

fibers that condense into bundles throughout the callus. In addition, osteoblasts deposit osteoid

along the collagen fibers, beginning the consolidation phase of DO. During the consolidation

phase, the fixator is left in place until the bone acquires enough strength to withstand mechanical

stress of weight bearing. The patient is usually asked to keep on the Ilizarov device on for a

month for every centimeter distracted. During consolidation, the process of bone remodeling

continues developing trabecular and cortical bone within the callus. The remodeled bone within

the distracted gap usually contains the same properties as the host’s original bone. [2-4]

20

21

Figure 2 : Phases of Distraction osteogenesis

Radiological images of a wild-type mouse model of DO demonstrate the process of bone regeneration within the distracted gap. Distracted tibial samples were collected from the different phases of DO.

21

1.1.4-Bone Development during DO

There are two types of ossification that are essential for osteogenesis: endochondral and

intramembranous ossification. Intramembranous ossification is a one-stage process in which

mesenchymal cell-derived osteoblasts produce bone. Endochondral ossification is a two-step

process that uses cartilaginous intermediate to develop new bones. [5] Both methods of

ossification are initiated by mesenchymal stem cell condensation. The condensed mesenchymal

cells that take part in intramembranous ossification develop into preosteoblast cells. [6] These

preosteoblast cells further differentiate into mature osteoblasts to take part in the bone

remodeling process, forming woven bone. [7] Contrarily, the multistage process of endochondral

ossification is initiated with mesenchymal condensation and the production of type II collagen.

Mesenchymal cell-derived chondrocytes undergo proliferation and perichondrial cells start

expressing type I collagen. [8] The type II collagen-expressing, proliferating chondrocytes form

a cartilaginous template. Eventually, the chondrocytes stop proliferating and become pre-

hypertrophic. The pre- hypertrophic chondrocytes mature into hypertrophic chondrocytes that

express type X collagen and participate in matrix mineralization. Following maturation, the

hypertrophic chondrocytes undergo cell death by apoptosis [9]. Periosteal-derived osteoblasts

and newly formed blood vessels invade the cartilaginous template for trabecular bone formation

eventually forming the primary spongiosa [10]. With continual bone remodeling by osteoblasts

and osteoclasts, the center of the primary spongiosa is split into two epiphyseal growth plates

that are pushed apart in opposite directions.[9]

22

1.1.5 Types of ossification in DO

Kojimoto and colleagues completed DO on young Japanese rabbits, stabilized by a unilateral

dynamic fixator. Their results concluded that the principal bone development process was

endochondral ossification. Their histologic studies detected the presence of elongated

cartilaginous cells and hypertrophic chondrocytes in the callus [11].

Delloye and colleagues performed DO on adult mongrel dogs using a circular external fixation

system. Intramembranous ossification was identified in their samples analysis. [12] Similarly,

the same conclusion was found by the group of Arnonson and colleagues. They performed DO

on a canine model using two types of fixators, the Ilizarov fixator and the Wagner fixator, and

also noticed intramembranous ossification was the only mechanism used for bone formation

during DO.[13]

However, Rauch and colleagues performed DO on New Zealand rabbits. The osteotomy was

completed on the right tibia of the rabbits and stabilized using Orthofix uniplanar fixators. They

found both types of ossification in their model.[14]

A group of Japanese researchers detected the presence of three types of ossification used within a

rat model of DO: endochondral, intramembranous and transchondroid ossification. Distraction

osteogenesis was performed on adult rats, in which bony fragments were supported by a

monolaternal external fixator. Histological findings showed the formation of a fibrocartilaginous-

based callus that became hypertrophic during early distraction, indicating endochondral

ossification was used during this time point of DO. By 10 to 20 days of distraction, histological

images revealed the presence of preosteoblasts, osteoblasts and fibroblast-like cells within the

23

cartilaginous callus. Eventually, the cartilaginous callus was resorbed and replaced with new

bone, demonstrating that intramembranous ossification was used during the late distraction phase

of DO. In between these two phases of bone development, transchondroid ossification was

observed during the mid-distraction phase of DO. During transchondroid bone formation,

chondrocyte-like cells produced chondroid bone, an intermediate type of tissue that contained

properties of both cartilage and bone. The chondroid matrix resembled bone matrix more than a

cartilaginous matrix. Furthermore, round chondrocyte-like cells and smaller osteocyte-like cells

were present in the histological sections of transchondroid bone. [15]

Finally, if we come back to the Principle of Tension and Stress, Ilizarov observed that type of

bone formation that occurred within a distracted gap depends on the stability of the external

fixation system. A stable fixation system ensures that the surrounding bone marrow and

periosteal tissues around the fracture site are intact, allowing bone regeneration to occur without

the development of cartilaginous intermediate. Thus, Ilizarov concluded that intramembranous

ossification is the main method of bone development used within a healthy callus of DO [2-4].

Contrarily, a cartilaginous callus may form within the distracted site via endochondral

ossification when there is an unstable fixation system. A cartilaginous callus is usually formed to

stabilize and support the fractured segments within the distracted gap.[16]

1.1.6-Complications and Drawbacks of DO

Like any surgery, DO comes with risks for different complications. The regular complications

related to the anesthesia can always apply. Then during the surgery itself, risks of bleeding,

injury to major vessels of nerves are always possible. In the post-operative period, the risk of

24

infections is always possible and complications related to the use of hardware occasionally

happened. Post-consolidation, there may be a possibility that the bone can refracture in the

distracted site or a different site during the removal of the external fixator. [17]

The main drawback associated with DO is the long period of time patients are required to keep

on the external fixation system on the affected site. [17] For every centimeter the bone is

lengthened, the external fixator has to been kept on for one month (i.e. if a patient requires ten

centimeters of lengthening, the fixator must kept on for ten months). This drawback can cause

social, psychological and medical problems for the patient and also lead to problems with patient

compliance. Thus, researchers are studying methods to accelerate the consolidation phase of DO.

1.2-Attempts to Accelerate DO

Many studies have been conducted using animal models to analyze the effects of physical and

molecular methods that can potentially accelerate the consolidation phase of DO. [17] Physical

methods have included the use of mechanical stimulation [18-19] and the application of low-

intensity pulse ultrasound (LIPUS) [20 to 24]. Whereas, biological methods have included the

transplantation of bone marrow [25] and osteoblastlike cells into the fracture site [26-27], the

local injection of bisphosphonates [28-29], and peptide growth factors.[30]

25

1.2.1-Mechanical Stimulation

There have been few attempts using mechanical stimulation, such as compressive stimulation, to

increase the consolidation phase of DO. The application of compressive stimulation during the

distraction phase of mandibular DO demonstrated no significant changes in bone regeneration

[31]. Kim et al. studied the effects of compressive stimulation during early consolidation in a rat

model of mandibular DO. DO was performed on the mandible of rats followed by a 3 day

latency period, a 4 day distraction period (a distraction rate of 0.25 mm/day twice daily) and a 7

day consolidation period. A compressive force was applied in the direction opposite to distraction

at the compression rate of 0.25 mm/day twice daily for the first four days of consolidation.

Therefore, after a total of 4.0 mm of distraction and 2.0 mm of compression, the total distraction

length was 2.0 mm. Controls were subjected to the same conditions of distraction in the absence

of compression. Histological and radiological data demonstrated the presence of a softer callus

that had primarily developed via intramembranous ossification in the experimental rat groups

compared to the control group during early consolidation. By late consolidation, the compressive

force helped decrease the size of the distracted gap, forming a thicker volume compact bone

regenerate. Thus, the use of compressive stimulation during the consolidation phase of

mandibular DO possibly contributed to the formation of more mature and dense bone by

accelerating the differentiation of osteoprogenitor cells to osteogenic cells.[19]

1.2.2- Low-Intensity Pulse Ultrasound (LIPUS)

Low-intensity pulsed ultrasound or (LIPUS) has also been studied for the acceleration of bone

healing during DO. LIPUS is a form of mechanical energy that travels to the site of bone repair

26

as pressure waves. At the fracture site, LIPUS enhances the process of angiogenesis, facilitating

the exchange of nutrients and waste removal among cells and stimulating the differentiation and

proliferation of fibroblasts, chondroblasts and osteoblasts. However, the exact mechanism of

LIPUS interaction with the surrounding soft tissue environment of the fracture site still remains

poorly understood. [20]

Sakurakichi and colleagues studied the application of LIPUS during the distraction phase of DO.

Distraction osteogenesis was performed on the right tibia of white Japanese rabbits using

unilateral external fixators (7 day latency period, 7 day distraction period using a distraction rate

of 1.5 mm/day and 7 day consolidation period). LIPUS was applied to the fracture site of rabbits

using an ultrasound transducer at 200µs burst of 1.5 MHz sine waves at a frequency of 1.0 kHz

for 20 minutes post- distraction each day (the same conditions are used in humans). The data

showed that application of LIPUS during the distraction phase of DO resulted in early bone

mineralization with high trabecular bone formation and increased mechanical strength of the

distracted rabbit tibia. [21] Similar results were observed in another rat model with unilateral

femoral lengthening and LIPUS application during the consolidation phase by Eberson group.

[22]

The application of LIPUS was also studied during the consolidation period. The Shimazaki’s

group performed DO using a unilateral fixator on the right tibia of rabbits (white Japanese). The

project was separated in this fashion : latency period (7 days), distraction (10 days) and

consolidation (21 days). The LIPUS was applied using the same conditions used in humans. The

27

results showed that the LIPUS treatment group compared to the controls had an increase in bone

mineral density and hard callus formation. [23] Another study also examining the effect of

LIPUS on consolidation came to similar conclusion. Claes and colleagues also analyzed the

outcomes of LIPUS stimulation on a sheep model of DO. Using a custom-made fixation system,

DO was performed on the metatarsus of the animals in this fashion : 4 days of latency, 16 days of

distraction and 64 days of consolidation. The sheep were stimulated with LIPUS for 20 minutes

each day starting the first day of consolidation. Results demonstrated a greater axial stiffness and

early callus formation in the LIPUS-treated sheep. In addition, a 32% increase in bone formation

was observed in the LIPUS-treated sheep compared to the controls that was predominantly

formed via endochondral ossification. This process of callus development in the LIPUS-treated

sheep is quite different from the LIPUS untreated controls that primarily form bone via

intramembranous bone formation. Thus, LIPUS treatment may enhance endochondral bone

formation within the distracted gap. [24]

1.2.3 Transplantation of Bone Marrow Cells (BMCs)

Transplantation of bone marrow cells (BMCs) has been studied in the recent years. They were

mainly used to shorten the consolidation period. The animal studies were created around the idea

that the transplantation of multipotent BMCs during DO would develop into osteogenic

progenitors may enhance bone formation within the fracture site. The advantages of BMC

transplantation include the easy isolation of these cells from bone marrow sections and it is a safe

treatment with relatively few side effects. [25] The biggest challenge with the use of BMC

28

transplantation is to find an appropriate delivery system at the distraction site. Thus, the choice of

osteoinductive factors and type of scaffold are important for BMC transplantation during DO.

Kitoh and colleagues discovered that the combination of culture expanded BMCs in platelet-rich

plasma (PRP) stimulated bone regeneration in patients that had undergone tibial or femoral

lengthening. PRP supplied osteoinductive factors, such as platelet-derived growth factor (PDGF),

insulin-like growth factor-1(IGF-1) and transforming growth factors (TGF-ßs) that are necessary

for bone regeneration. Furthermore, the use of a biodegradable scaffold helped prevent infection

and inflammation of the fracture site.[32]

Kitoh’s study measured a standardized healing index for each patient. This healing index was

calculated by the division of the time required for complete consolidation by the total distracted

length observed by x-ray analysis. BMC-PRP-treated patients had a smaller healing index than

untreated patients, demonstrating a decrease in the consolidation time for DO. Furthermore, a

delay in consolidation was observed in 45% of untreated BMC-PRP patients compared to the

BMC-PRP-treated patients that exhibited normal bone healing patterns. [32]

1.2.4- Transplantation of Osteoblast-like cells

Transplantation of periosteum-derived osteoblast-like cells were also studied during DO. Tsubota

and colleagues studied the effects this technique in a white Japanese rabbits using a hemilateral

external fixator. The treatment groups were divided in 3 : group 1- a control group that

underwent distraction without the transplantation of osteoblastlike cells, group 2- a control group

of rabbits that were only injected with physiological saline and group 3-test group of rabbits that

29

were injected with periosteum-derived osteoblast-like cells dissolved in physiogical saline. The

rabbits underwent 7 day latency period, 20 day distraction period with a distraction rate of 1mm/

day and sacrifices were made every 2 weeks during consolidation. Results showed that group 3

rabbits contained larger and stronger callus formation than the controls as measured by a high

transaxial area ratio and three point-bending analysis. Furthermore, the bone mineral density of

the treatment group was significantly higher than the control groups at all time points. [26]

Tamakine et al. studied in 2002 the use of bone marrow-derived osteoblast-like cells using a

collagen-based scaffold in a rat model. DO was performed on the left femur of rats and stabilized

using a unilateral fixator. Post-surgery, rats were subjected to a 7 day latency period and a 10 day

distraction period with a distraction rate of 0.25 mm every 12 hours. The treatment group was

receiving an injection of osteoblast-like cells in collagen gel in the distraction zone. The control

groups injected with : physiological saline, collagen gel or bone marrow derived osteoblast-like

cells without the gel vector. The results showed that the rats from the treatment group had larger

callus formation compared to the controls. The group of rats that were treated with bone marrow-

derived osteoblast-like cells in gel also contained an increase in fracture strength two, four and

six weeks post- transplantation as determined by three-point bending analysis. Therefore, both

studies demonstrated that the transplantation of osteoblast-like cells enhanced bone formation in

the rabbit and rat models of DO. It is possible that the transplantation of the osteoblast-like cells

population into the distracted zone increased the number of osteogenic cells and recruited

osteoinductive factors to the fracture site that are necessary for bone regeneration, that may

account for the large callus formation [27]

30

1.2.5-Demineralized Bone Matrix

Demineralized bone matrix has been used in the past for the treatment of non-unions [33], spinal

fusions [34], craniomaxillofacial defects[35-36], and segmental bone defects [37-39]. The first

ones to investigate the effect of demineralized bone matrix during DO were Hagino et al. in 1999

[40]. Their study was constructed around the use of grafting demineralized bone matrix during

lengthening of the tibia in young Japanese White rabbits. The demineralized bone matrix was

made from frozen cortical bone harvested from other rabbits. The control group was not

receiving any graft during the process. Their study was able to show a significant difference in

the formation of a radiographic callus and in the rate of non-union favoring the grafted group.

They also tried to accelerate their distraction rate up to 2-3 mm/day and still got acceptable

results. Thus, they demonstrated that the use of demineralized bone matrix can allow for

satisfactory bone formation at faster rates than normal.

1.2.6- Bone-graft substitutes

Synthetic or natural biomaterials have been used as bone-graft substitutes to autologous or

allogeneic bone grafts. Although they lack osteoinductive or osteogenic properties, synthetic

bone substitutes and biomaterials are already widely used in clinical practice for

osteoconduction. They consist of scaffolds that promote the migration, proliferation and

differentiation of bone cells for bone regeneration. A wide range of synthetic bone substitutes

are currently used. The main ones currently used being collagen, hydroxyapatite (HA), β-

tricalcium phosphate (β-TCP), calcium-phosphate cements, and glass ceramics [41,42].

31

Especially for reconstruction of large bone defects, for which there is a need for a substantial

structural scaffold, an alternative to massive cortical auto or allografts is the use of cylindrical

metallic or titanium mesh cages as a scaffold combined with cancellous bone allograft,

demineralized bone matrix or autologous bone [43,44]. Furthermore, there are also non-

biological osteoconductive substrates, such as fabricated biocompatible metals (for example,

porous tantalum) that offer the potential for absolute control of the final structure without any

immunogenicity [41].

Wang and al. showed in 2008 in a study comparing 3 groups of rabbits with a 1.0 cm bone defect

on their left tibia. Group A, the defect gap was reduced with the tibia shortened for 1.0-cm and

received lengthening for 10 days. Group B, the defect gap was filled with 1.0-cm restorable

porous hydroxyapatite and Tri-calcium phosphates cylindrical block (HA/TCP block, diameter is

0.5-cm) and did not receive lengthening. Group C, The 1.0-cm defect gap was reduced 0.5 cm

and the remaining 0.5-cm defect gap was filled with the 0.5-cm HA/TCP block and received

lengthening for 5 days. The bone specimens were subject to microcomputed tomography,

mechanical testing, and histological examinations. Their results showed that a combined use of

biomaterials and DO technique can reduce the treatment time and enhance bone consolidation in

bone defect management. [45]

Research is ongoing to improve the mechanical properties and biocompatibility of scaffolds, to

promote osteoblast adhesion, growth and differentiation, and finally to allow vascular ingrowth

and bone-tissue formation. Improved biodegradable and bioactive three-dimensional porous

scaffolds [46] are being investigated, as well as new technologies using nanotechnology, such as

magnetic biohybrid porous scaffolds acting as a crosslinking agent for collagen for bone

32

regeneration guided by an external magnetic field [47], or injectable scaffolds which provide

easier application [48].

1.2.7-Bisphosphonates

In theory the biphosphonates act by decreasing the catabolic activity during DO. Takahashi and

colleagues studied the effects of administering nitrogen-containing bisphosphonates on a rabbit

model of DO. Rabbits underwent tibial lengthening with a 5 day latency period, 21 day

distraction period (distraction rate of 0.35mm every 12 hours) and a consolidation phase of four

weeks. Rabbits were injected with 0.4 mg/kg of nitrogen-containing bisphosphonates for a period

of 6 weeks post-osteotomy. Controls were injected with vehicle only. Results showed that the

bisphosphonate-treated rabbits contained new bone with greater mechanical strength in the

osteopenic zones including 5.6x greater bone volume and 3.3x greater mechanical resistance than

the controls.[29]

33

1.2.8-Growth Factors

The use of different growth factors may potentially be the most resourceful technique for

accelerated bone healing during DO. Different growth factors have been analyzed in fracture

healing studies including Growth Hormone (GH) Transforming growth factor ß (TGF- ß),

Platelet-derived growth factor (PDGF), Insulin-like growth factors (IGF), Fibroblast growth

factor (FGF) and Bone morphogenetic proteins (BMP). [30]

1.2.8.1-Growth Hormone Injection

Growth hormone injections was studied by Bail et al. in 2002. They applied this treatment to the

process of mandibular DO. They demonstrated that systemic GH administration significantly

increases the torsional stability of the regenerate in comparison to the contralateral side with no

exogenous GH administration and showed a larger callus formation. [49]

1.2.9-Cartilage Oligomeric Matrix Protein - angiopoietin-1

(COMP-Ang1)

COMP-Ang1 is an angiogenic factor that has been studied in DO. Park et al. in 2010 investigated

the effect of local COMP-Ang1 administration in the center of the distraction zone [50].Control

rats underwent tibial distraction without treatment. In the two remaining groups, BSA (100 ug) or

COMP-Ang1 (100 ug) were injected transcutaneously into the center of the distraction zone.

34

They demonstrated that bone formation in the distraction gap progressed more quickly in the

COMP–Ang1-treated group than in the BSA-treated group. Also, histological findings and

immunostaining of endothelial cells for factor VIII revealed that the COMP–Ang1 group of

animals exhibited higher levels of vascularity. NanoCT and dual-energy X-ray absorptiometry

analysis also revealed increased new bone formation along capillaries in the COMP–Ang1 group

compared with the BSA group show an increase. Runt-related transcription factor 2 and its target

genes, including bone sialoprotein, type 1 collagen, osteopontin, and osterix were significantly

upregulated in the COMP–Ang1 group. Their results suggested that COMP–Ang1 could be

potentially used as a therapeutic agent for treatment of distracted limbs by enhancing

angiogenesis [50].

1.2.10-Bone Morphogenetic Proteins (BMPs)

BMPs had been involved in the regulation of cell proliferation, survival, differentiation and

apoptosis. They play a pivotal role in inducing bone, cartilage, ligament, and tendon formation at

both heterotopic and orthotopic sites. 91 BMPs were first identified by Dr. Marshall Urist in

1965 when he noticed at implantation of demineralized bone matrix in different sites of a rodent

model induced ectopic bone formation.[50] Twenty BMPs have been identified to date and

except for BMP1, all BMPs are a part of the transforming growth factor ß (TGF- ß) superfamily.

[51]

BMPs are known to be potent inducers of osteogenesis during embryological bone formation,

and fracture healing. These proteins act through serine-threonine kinase transmembrane

35

receptors, type I and type II [52-53]. Their activities are mediated through Smads signaling

pathways.

1.2.10.1-BMPs Function

BMPs have been implicated in a variety of functions including bone and cartilage generation and

a number of non-osteogenic roles.

BMPs have also been shown to be important regulators of chondrogenesis and skeletogenesis

during normal embryological development.

BMP-2, -4, -6, -7, and -9 have the greatest osteogenic capacity. BMP-2 has been demonstrated to

be expressed strongly around cartilage condensations, periosteal and osteogenic zones. BMP-4

on the other hand is expressed in the perichondrium and BMP-6 is expressed in hypertrophic

chondrocytes. BMP-7 has been observed to be expressed highly in the perichondrium with no

expression in the zones of joint formation [51].

Both BMP-2 and -7 have been implicated to have active critical roles in the expression of major

transcription factors during osteogenesis such as Runt-related transcription factor 2 (Runx2 ) and

Osterix (Osx) in mesenchymal stem cells. BMP-7 knock-outs exhibit skeletal alterations in the

rib cage, hind limbs and skull. [54]

BMPs appear to play an important role in regulating stem cells differentiation. In embryonic

stem cells, BMP signaling appears to be required for self-renewal. In mesenchymal stem cells,

36

BMP signaling stimulates osteoblastic differentiation. However, it has been suggested that due to

the pleiotropic functions of BMPs, it is possible for the signaling to be mutagenic or oncogenic.

In particular, BMP-7 located at 20q13, has been demonstrated to play a key role in osteoblast

differentiation. [51]

1.2.10.2-BMP Pathway

BMP ligands bind to two distinct receptors: type I and type II serine/threonine kinase receptors.

Both of these receptors are required for signaling [119]. There are three type I receptors: type IA

and IB BMP receptors (BMPIA or ALK3 and BMPIB or ALK6), type IA activin receptor

(ActRIA or ALK2). Type II receptors also exist in three forms: type II BMP receptor (BMPR-II),

type II and IIB activin receptors (ActR-II and ActR- IIB). [55]

BMP ligands bind to type II receptors. The serine/threonine kinase domains of type II receptors

are constitutively active and phosphorylate Gly-Ser [50] domains of type I receptors upon ligand

binding, leading to activation of type I receptor kinases [55]. This leads to the recruitment of

pathway restricted Smads (R-Smads, Smads1, 5 or 8) and later recruitment of the common

mediator Smad (Co-Smad, Smad 4) into the complex. This complex migrates into the nucleus

and activates the transcription of specific target genes [57].

The signaling of BMP is modulated at different steps along the pathway. In the extracellular

environment, antagonists such as Cerbarus, Dan, Gremlin, Noggin, and Inhibin which are known

to bind specifically to BMP receptors can regulate initiation of the cascade. Additionally at the

37

cell surface, the oligomerization mode of the receptors can activate the signaling pathway

specifically [51].

Once the signal is transduced into the intracellular compartment, the signal can be modulated by

the activation of inhibitory Smad proteins (I-Smad, Smad 6, 7) or Smurf 1. In the nucleus, there

are a number of co-activators needed for the activation of specific target genes and their

transcription can be inhibited by co-repressors [51, 58].

38

nal induces osteoblastic di!erentiation through Bmpr1b butinhibits osteoblastic di!erentiation through Bmpr1a [8]; inDrosophila germline stem cells (GSCs), Dpp (homolog ofBMP2/4) is essential for the maintenance of stem cells [9].

BMPs may have pleiotropic functions including mutagene-sis or oncogenesis, thus, extensive studies have beenreported in the field of musculoskeletal oncology, especiallyosteosaroma [10]. Urist et al. first reported that certain via-ble and devitalized human osteosarcomas induced ectopicbone formation in athymic nude mice, suggesting thathuman osteosarcomas may produce BMP-like proteins. Insubsequently studies, BMP-2/4 proteins or genes in a vari-ety of bone tumors have been studied by immunohisto-chemistry, in situ hybridization, or both [11]. BMPscontrol many key steps in the formation and di!erentiationof the vertebrate nervous system, which act at di!erentstages of neural development and in di!erent regions ofthe CNS to regulate cell fate, proliferation and di!erentia-tion [12]. BMP-2, BMP-4, and BMP-6 have been localizedto areas of vascular calcification. Subsequently, studiesshowed that BMP-2 inhibiting vascular smooth muscle cellsproliferation when stimulated with serum or growth factorsin vitro [13]. During embryonic development, BMPs medi-ate programmed cell death, or apoptosis, the process thatremoves unnecessary tissues, thereby ensuring proper mor-phogenesis. BMP-4 was first demonstrated to induce apop-tosis in rhombomeric neural crest cells [14]. The function ofBMPs family members is shown in Table 1.

Receptors and molecular signaling

Members of the BMP family bind to two distinct type IIand type I serine/threonine kinase receptors, both of whichare required for signal transduction [15]. There are threetype II receptors have been shown to bind BMP ligands:type II BMP receptor (BMPR-II), type II and IIB activinreceptors (ActR-II and ActR-IIB), and three type I recep-tors for BMPs have also been characterized: type IA andIB BMP receptors (BMPIA or ALK3 and BMPIB orALK6), type IA activin receptor (ActRIA or ALK2)

Table 1The list of BMP family members

BMP Gene locus Function Receptors and inhibitors

BMP-1 8p21 It is a metalloprotease that acts on procollagen I, II, and III. It isinvolved in cartilage development

Type I receptors

BMP-2 20pl2 Acts as a disulfide-linked homodimer and induces bone and cartilageformation. It is a candidate as a retinoid mediator. Plays a key role inosteoblast di!erentiation

ACVRIA, ACVRIB, ACVRIC,ACVRLI-BMPR (BMPRIA–BMPRIB)

BMP-3 14p22 Induces bone formation Type II receptorsBMP-4 14q22–q23 Regulates the formation of teeth, limbs and bone from mesoderm. It

also plays a role in fracture repairACVR2A, ACVR2B, AMHR2,BMPR2

BMP-5 6p12.1 Performs functions in cartilage developmentBMP-6 6p12.1 Plays a key role in joint integrity in adults Type III receptorsBMP-7 20q13 Plays a key role in osteoblast di!erentiation. It also induces the

production of SMADI. Also key in renal development and repairBetaglycan

BMP-8 1p35–p32 Involved in bone and cartilage developmentBMP-9 Promoting chondrogenic di!erentiation of human multipotential

mesenchymal cellsLigand inhibitors

BMP-10 2p14 Play a role in the trabeculation of the embryonic heart Cerberus, Chordin, DAN, Decorin,Follistatin, Gremlin, Lefty, LTBPI

BMP-15 Xp11.2 May play a role in oocyte and follicular development Noggin, THBSI

In this table, it shows the function (if known), gene locus, receptors and ligand inhibitors.

Fig. 2. Bone morphogenetic protein (BMP) receptors mediate BMPsignaling through activating Smad. BBMP ligands bind to the BMPreceptors BMPRI and BMPRII. After BMPRII phosphorylated, follow-ing activates BMPRI. Phosphorylated BMPRI subsequently phosphory-lates receptor-activated Smad proteins (R-Smads), which associate withcommon mediator-Smad (co-Smad) and enter the nucleus, where theyregulate gene expression. The Smad proteins regulate promoter activity byinteracting with transcriptional co-activators or co-repressors to positivelyor negatively control gene expression. The BMP signal can be blocked byextracellular antagonists, such as noggin, which bind BMP ligands andprevent their association with the BMP receptors, as well as byintracellular proteins, such as inhibitory Smads (I-Smads), which preventthe association between R-Smads and co-Smad and Smurf 1. Smurf1 is aHect domain E3 ubiquitin ligase. It interacts with Smad1 and 5 andmediates the degradation of these and also forms a complex with Smad6,is exported from the nucleus and targeted to the type I BMP receptors fortheir degradation [21]. Modified from Niswander et al.

552 Y.-T. Xiao et al. / Biochemical and Biophysical Research Communications 362 (2007) 550–553

Figure 3 : BMP pathway

Bone morphogenetic protein (BMP) receptors mediate BMP signaling through activating Smad. BBMP ligands bind to the BMP receptors BMPRI and BMPRII. After BMPRII phosphorylated, following activates BMPRI. Phosphorylated BMPRI (phosphorylation expressed has p-bodies in figure) subsequently phosphorylates receptor-activated Smad proteins (R-Smads), which associate with common mediator-Smad (co-Smad) and enter the nucleus, where they regulate gene expression. The Smad proteins regulate promoter activity by interacting with transcriptional co-activators or co-repressors to positively or negatively control gene expression. The BMP signal can be blocked by extracellular antagonists, such as noggin, which bind BMP ligands and prevent their association with the BMP receptors, as well as by intracellular proteins, such as inhibitory Smads (I-Smads), which prevent the association between R-Smads and co-Smad and Smurf 1. Smurf1 is a Hect domain E3 ubiquitin ligase. It interacts with Smad1 and 5 and mediates the degradation of these and also forms a complex with Smad6, is exported from the nucleus and targeted to the type I BMP receptors for their degradation [62] . Modified from Niswander et al. [51]

39

1.2.10.3 Use of exogenous BMPs during Distraction Osteogenesis

Usage of exogenous BMPs during distraction osteogenesis have been tested as a treatment to

accelerate the process of DO. Mizumoto et al. have studied in 2003 the use of recombinant

human BMP-7 during DO in a rat model. The rats were divided in 2 groups : one group was

treated with rhBMP-7 in an aquaeous solvent and the other group just received the solvent

(control) through an injection in the osteotomy site. Their results showed an increase in bone-

mineral density and biomechanical testing results in the treatment group.[60] Yang and al studied

in 2012 the use of BMP-2 during DO through a tri-calcium phosphate (TCP)/hydroxyapatite

(HA) delivery system in a rat model. The animals were randomized into three groups: group I

served as a control, group II was treated with only TCP/HA, and group III was treated with

recombinant human (rh) BMP-2-coated TCP/HA. Materials were inserted into the medullary

canal at the femoral osteotomy site at the end of the lengthening period. Their results showed that

application of rhBMP-2, at the end of the rather rapid distraction period, as a single bolus

significantly increased the osteogenic process, while beta-TCP/HA behaved effectively as a

sustained delivery system for this osteoinductive protein. [61]

Studies investigating the clinical effects of BMP administration in human long bone fractures

have also been numerous during the past decade. A Cochrane review was published in 2010

studying eleven randomized control trials. Their analysis showed that apart from one study, the

times to fracture healing were comparable between the BMP and control groups. There was some

evidence for increased healing rates, without requiring a secondary procedure, of BMP compared

40

with usual care control in acute, mainly open, tibial fractures (risk ratio (RR) 1.19, 95% CI 0.99

to 1.43). The pooled RR for achieving union for nonunited fractures was 1.02 (95% CI 0.90 to

1.15). One study found no difference in union for patients who had corrective osteotomy for

radial malunions. Data from three RCTs indicated that fewer secondary procedures were required

for acute fracture patients treated with BMP versus controls (RR 0.65, 95% CI 0.50 to 0.83).

[63]

1.2.10.3.1 Complications usage of exogenous BMPs

Some controversies linger around the usage of BMPs. Even if some studies have demonstrated

that therapy with rhBMP-7 in non-unions is significantly more cost-effective than traditional

treatment modalities [65], most of the recent systematic reviews come with the opposite

conclusion. [63-68] Even if multiple studies have also demonstrated that BMP-2 and -7 are

generally safe, effective, and at times superior alternatives to autologous bone grafting during

interbody spinal fusion procedures [65-66], some animal studies have reported some

complications. Recently Zara and Al. studied the use of high doses of BMPs-2 in a rat model.

Their results showed that they consistently reproduced BMP2 side effects of cyst-like bone and

soft tissue swelling using high BMP2 concentration approaching 1500 µg/mL. [67]

41

1.2.11 BMP Antagonists

BMP antagonists have been shown to modulate BMP activities in diverse and critical ways in a

myriad of tissues and systems during normal vertebrate development. [69]

There are three main types of BMP antagonists: pro- region inhibitors, ligand antagonists, and

receptor antagonists [70].

Pro-region inhibitors interact with the mature peptide [70]. Recent studies with BMP-2 and -9

have shown that the pro-region inhibitors are inefficient inhibitors of osteogenesis as they can

easily get degraded by proteolytic cleavage [71-72].

The second group of inhibitors is the ligand antagonists such as Noggin, Chordin, Follistatin,

Gremlin, and Sclerosin that bind directly to the BMP ligand and prevent the interaction of the

complex with the receptor.

Lastly, the receptor antagonists are the group of inhibitors that interact with the BMP receptors

and prevent the BMP-receptor interaction. Additionally, inhibitory Smads (Smad 6 and Smad 7)

are the BMP antagonists that interfere with BMP activity at intracellular level [70].

1.2.11.1 Noggin

Noggin is a potent inhibitor of BMP activity that directly binds BMP in the extracellular

environment. Noggin binds with differing affinities to BMP-2, -4, -5, -6, and -7, with the highest

affinities observed between noggin and BMP-2 and -4 and the relatively weakest affinity

42

between noggin and BMP-7 [73-74]. Noggin is essential for normal skeletal and joint

development [75-76]. A single dose of noggin in rats significantly decreases the rate of

membranous ossification as measured by a bone in-growth chamber technique. Interestingly, the

total amount of tissue in-growth was similar between controls and the noggin groups, but

ossification was significantly reduced in the noggin group [73].

Noggin expression is clearly upregulated by BMP as a protective feedback mechanism to prevent

bone overgrowth in BMP stimulated osteoblasts [77].

The laboratory of Wan et al. [78] realized in vitro and in vivo studies using siRNA to silence

noggin activity . Pre-osteoblasts were infected with specific siRNA constructs that were shown

to effectively silence noggin expression. In these infected cells, expression of Smad 1 and 5, and

rate of osteoblast differentiation were all significantly increased compared to sham infected

controls. Expression of osteoblastic differentiation markers Runx2, osteocalcin, and osteopontin

all peaked approximately three days earlier in the siRNA infected cells compared to sham

infected controls. For their in vivo studies, defects were created in mouse calvaria and the

cortical defects were seeded with either siRNA infected osteoblasts or sham infected osteoblasts.

At two and four weeks, the defects seeded with siRNA infected osteoblasts demonstrated

significantly more bone formation. Interestingly, at eight weeks there were similar levels of bone

formation with no appreciable difference between siRNA infected and sham infected control

groups.

Their results were suggesting that it may be possible to manipulate noggin modulation of BMP

activity in such a way to enhance and speed bone regeneration and, potentially, fracture healing.

43

Studies examining the effects of noggin silencing on long bone fracture healing have yet to be

performed, but are necessary to further the possibility of therapeutic algorithms utilizing the

manipulation of noggin. Noggin expression peaks in the callus 10 days post fracture [79].

1.2.11.2 Chordin

Chordin appears to bind BMP-2, BMP-4, and BMP-7 in the extracellular environment similar to

noggin and thus prevents BMP activity by preventing it from binding to receptors [80-83].

Chordin is very important in embryological development, especially skeletogenesis [81-83].

Zhang et al found in 2002 that chordin expression during development was shown to be

inversely related to the stages of chondrocyte maturation, indicating that chordin is likely an

inhibitor in this process [81]. Then Nilsson et al. found that chordin expression is significantly

lower in the proliferative and hypertrophic zones versus the resting zone in 7-day old rat tibial

physes. [84] Despite conclusive evidence that chordin is an inhibitor of BMP during normal

development, few studies to date have specifically investigated whether chordin plays any role

during fracture healing. One investigation demonstrated a peak of chordin expression within

callus 10 days after fracture in normally healing fractures and a consistent level of chordin

expression in experimentally produced non-unions [79].

44

1.2.12 RNA interference (RNAi)

RNA interference (RNAi), an accurate and potent gene-silencing method, was first

experimentally documented in 1998 in Caenorhabditis elegans by Fire et al., who subsequently

were awarded the 2006 Nobel Prize in Physiology/Medicine. [85]

RNAi is a natural process in which the expression of a targeted gene can be knocked down with

a good specificity and selectivity. Methods of mediating the RNAi effect involve small

interfering RNA (siRNA), short hairpin RNA (shRNA) and bi-functional shRNA. The simplicity

of siRNA manufacturing and transient nature of the effect per dose are optimally suited for

certain medical disorders (i.e. viral injections). However, using the endogenous processing

machinery, optimized shRNA constructs allow for high potency and sustainable effects using low

copy numbers resulting in less off-target effects. Bi-functional design may further enhance

potency and safety of RNAi-based therapeutics. [86]

45

1.2.12.1 shRNA

shRNAs are synthesized in the nucleus of cells, further processed and transported to the

cytoplasm, and then incorporated into the RISC for activity [87]. To be effective, the shRNA are

designed to follow the rules predicated by the specifics of the cellular machinery.

shRNA can be transcribed by either RNA polymerase II or III through RNA polymerase II or III

promoters on the expression cassette. The primary transcript generated from RNA polymerase II

promoter contains a hairpin like stem-loop structure that is processed in the nucleus by a

complex containing the RNase III enzyme Drosha and the double-stranded RNA-binding domain

protein DGCR8 [88]. The complex measures the hairpin and allows precise processing of the

long primary transcripts into individual shRNAs with a 2 nt 3′ overhang [89]. The processed

primary transcript is the pre-shRNA molecule. It is transported to the cytoplasm by exportin 5, a

Ran-GTP- dependent mechanism [90-91]. In the cytoplasm the pre-shRNA is loaded onto

another RNase III complex containing the RNase III enzyme Dicer and TRBP/PACT where the

loop of the hairpin is processed off to form a double-stranded siRNA with 2 nt 3′ overhangs

[92-94]. The Dicer containing complex then coordinates loading onto the Ago2 protein

containing RISC as described earlier for siRNA. Pre- shRNA has been found to be part of the

RLC; thus, pre-shRNA may potentially directly associate with RLC rather than through a two

steps process via a different Dicer/TRBP/PACT complex [95].

46

Figure 4 : shRNA mediated RNA interference pathway

Schematic of the shRNA mediated RNA interference pathway. After delivery of the shRNA expression vector into the cytoplasm, the vector needs to be transported into the nucleus for transcription. The primary transcripts (pre-shRNA) follow a similar route as discovered for the primary transcripts of microRNA. The primary transcripts are processed by the Drosha/DGCR8 complex and form pre shRNAs. Pre-shRNAs are transported to the cytoplasm via exportin 5, to be loaded onto the Dicer/TRBP/PACT complex where they are further processed to mature shRNA. Mature shRNA in the Dicer/TRBP/PACT complex are associated with Argonaute protein containing RISC and provide RNA interference function either through mRNA cleavage and degradation, or through translational suppression via p-bodies. [96]

47

1.2.12.2 Delivery systems shRNA

Efficacy of an RNAi therapeutic is limited by the quantity of the oligomer that effectively enters

the cells. In the clinical setting this is primarily dependent on the method of delivery.

Viral vectors are popular for laboratory delivery of shRNA because of their high transfection

efficiency and effective integration of exogenous DNA, but they have been losing support in

recent years because of concerns over safety and immunogenicity [96-97]. Non-viral polymeric

delivery systems, in particular those with biodegradable components, have much better safety

profiles than their viral counterparts though their transfection efficiency is generally lower.[98 to

100].

There are three major classes of non-viral delivery vehicle systems: synthetic polymers, natural/

biodegradable polymers, and lipids; many of the vehicles that are showing promise are actually

hybrids of these classes.

1.2.12.2.1 Lentiviral vector

Lentiviral vectors is an attractive gene delivery system for stable gene transduction in relatively

quiescent cells. This is why, lentiviral vectors have also been used for direct in situ infection

within a living organism. [101-103]

48

Unlike other retroviruses, lentivectors do not necessarily require cell division for proviral

integration and productive infection [104] and so offer a gene delivery method that does not need

growth factor administration to induce cell proliferation in cells transduced ex vivo. Additionally,

lentivectors are sufficiently robust for in vivo administration. Dong Sung An and Al. reported in

2003 the successful construction of lentiviral vectors to express shRNA stably in human cells.

They demonstrated that lentiviral vectors expressing siRNA directed to the reporter gene

luciferase, when stably transduced into human cells without drug selection, were capable of

protecting the cells from infection by a lentiviral vector encoding humanized firefly luciferase as

a reporter gene. [102]

49

2- OBJECTIVES OF STUDY

The objectives of the study if a successful knockdown of Noggin and Chordin would promote or

accelerate osteogenesis. The first objective or our study was to do a successful knockdown of

Noggin and Chordin in an in vitro setting using shRNAs through a lentiviral delivery system.

Then the second objective was to evaluate if an injection of those shRNAs through a lentiviral

delivery system would increase osteogenesis in an animal model of DO (wild-type mice).

50

3- MATERIALS & METHODS IN VITRO STUDIES

First RT-PCR were realized on non-transfected culture of MC3T3-E1 to measure the mRNA

level of Noggin and Chordin expression at various timepoints. The purpose was to identify the

timepoints of highest expression.

MC3T3-E1 cells were transduced with lentiviruses expressing various shRNAs targeting the

mouse Noggin (5 shRNAs) and Chordin (3 shRNAs) genes and 1 negative control.(Sigma-

Aldrich). On day 7 after infection, levels of RNA expression for Noggin and Chordin were

monitored through RT-PCR. Western blotting were performed on the cell extracts and culture

media to verify the expression and secretion of Noggin and Chordin proteins. Cell extracts were

also analyzed on day 2 and 4 after transduction for alkaline phosphastase activity which is a

marker of osteogenic differentiation.

3.1- shRNA selection

Using the Sigma-Aldrich company selection, 5 shRNAs targeting Noggin, 3 shRNA targeting

Chordin were selected. A control was also chosen : NT which is a non-target shRNA.

51

noggin NM 008711 (699bp)

3 4 5 1 2

chordin NM 009893 (3290bp)

1

3 2

Figure 5 : Position of shRNAs in cDNA Position in cDNA of different sequences of shRNAs used in our studies

GD218 (GD206-CMV-GFP)8249 bp

ampi

part of Env GP160 glycoprotein

rat Apin (no tag)

GFPPPT

partial U3

SIN/3'-LTRR

U5'SV40pA

pUCori

HIV 5'-LTRSDPsi

5'GAG D3rdG

RRE

pLKO-REV

pLKO-FWD

RSV CMV

bla promoter

EcoRI (1180 )

Mfe I (7399)

Figure 6 : Lentiviral plasmid Lentiviral plasmid used for our transduction of the different shRNAs studied in our project.

52

3.2 - Lentiviruses production

HEK293 were used for the first transfection. HEK 293 is a cell line originally derived from

human embryonic kidney cells. They have been used a lot in cellular biology because for their

easy growth, transfection and production of proteins and viruses. On day 1, the HEK293 were

seeded. The next day, they were transfected with the lentiviral shRNA plasmids using FuGENE 6

(Roche Applied Science) reagent following a ration of 3:1 (volume FuGENE : ug of DNA).

FuGENE 6 reagent was added to to media without serum and then vortex for 1 second. The mix

was then incubated at room temperature for 5 minutes. The plasmids containing the different

shRNAs where added to the mix of FuGENE and media and incubated at room temperature for

15 minutes. The plamids FuGENE mix was transfected into HEK293 cells. Culture were grown

for a period of 96 hours and the media containing the lentiviruses were collected and filtered

through 0.45 um filters. The lentiviruses were concentrated by ultracentrifugation at 25 000 rpm

for 2.5 hours. The viral pellet were resuspended in 100 ul of PBS, aliquoted, and frozen at -80 C.

[106]

3.3 - MC3T3-E1 transduction

MC3T3-E1-E1 cell line is a cell line that behave as immature, committed osteoblast cells. [107]

The lentiviruses produced in the earlier step were added to MC3T3-E1 cells in the presence of

polybrene (8ug/ ml) in 6 well-plates. After 24 h, the media was changed and the cells were

grown to confluence. The cells were then passage into 10 cm dishes and Puromycin (8ug/ml)

53

was added to select stable pools of stably expressing clones. As a control wild-type MC3T3-E1

were also treated with puromycin to monitor for selection efficacy.

3.4 - RT-PCR study

Then we decided to see if a successful knockdown of Chordin and Noggin had been produced by

our transduction. We first went ahead with PCR studies.

3.4.1 - RNA extraction from cell cultures

Every cultures were first washed 2-3 times using PBS. Then TRIzol (Invitrogen) was added to

the cells (at least 1ml/10cm2). Multiple up and down were realized using the pipet to assure a

good lysation of the cells. The cellular lysate were then stored at -20C.

3.4.2 - RNA purification

For each cellular lysate, 200ul of chlorophorm per ml of TRIzol was added. After obtaining a

good homogenization and being incubated for 3 minutes at room temperature, the samples were

centrifuged at 10 000 rpm for 15 mins at 4C. The centrifugation permitted to recuperate the

supernatant that was collected in new eppendorf tubes. A volume of 500 µl of isopropanol was

added to the samples to precipitate the RNA. The samples were mixed by inversion and allowed

to stand at room temperature for 10 minutes. The samples were centrifuged at 12 000 rpm for 10

minutes at 4C. The RNA pellets were washed with 1 ml of cold 75% ethanol per ml of TRIzol.

(Invitrogen) Again the mix was spun down at 12 000 rpm for 5 minutes at 4C. After removal of

ethanol, the samples were allowed to dry at room temperature for 30 minutes. The RNA pellets

were resuspended in DEPC water.

54

3.4.3 - RNA Quantification

We quantify the RNA using UV (OD260/ OD280). The RNA was diluted in sterile H20 and a

calculation of the quantification was made using this equation : µg/µl = (OD260 x Dilution factor

x 40)/1000.

3.4.4 - Reverse transcription of RNA

We used the applied Biosystems High-Capacity cDNA Reverse transcription kit. First the RNA

was diluted to a concentration of 2ug in 10 uL of DEPC H2O. Then a Master Mix of 2uL of 10x

RT buffer, 0.8uL of 25X dNTP Mix (100 mM), 2 uL of 10X RT Random primers, 1.0L of

MultiScribe Reverse Transcriptase and 4.2 uL of nuclease-free H20 were mixed. After, 10 uL of

diluted RNA and 10 uL of the Master Mix were mixed together in a well late and used for RT-

PCR. The GeneAmp PCR System 9700 thermal cycler (Applied Systems) was programmed as

followed : 10 mins at25C, 120 mins at 37C, 5 mins at 85C and then 4C until recuperated from

the machine.

3.4.5 - Real-Time PCR

Reverse transcribed mRNA collected were loaded into a 96-well plate and mixed with Universal

Master Mix (Applied Biosystems), specific TaqMan probe for Noggin and Chordin (TaqMan®

Gene Expression-Applied Biosystems) and RNAse free water. The 96-well plate was placed into

55

a 7500 Real Time PCR system (Applied Biosystems) for RT-qPCR analysis. Actine was used as

an endogenous control to normalize all samples used in the RT-qPCR reaction. Relative

quantification of the target RNA was performed using Applied Biosystems’ comparative Ct

method (ABI Prism 7700 Sequence Detection System User Bulletin #2, 2001).

3.5 - Western Blot study

3.5.1 - Protein collection from cell cultures and media

The stably expressing pools of MC3T3-E1 cells were seeded and grown to confluence in 6 well-

plates. The media was then switched to serum-free media and the cells were further grown for

48h. Then, the media and the cells were collected for downstream analysis.

The media was collected in eppendorf tubes and centrifugated for 5 mins. The supernatant was

then collected and Thricholoroacetic acid was added in a final concentration of 10% (v/v). The

mix was putted on ice for 30 mins. Then we proceeded with centrifugation again at 13 000 rpm.

The pellets were recuperated and washed with -20C acetone and centrifugated again at 13 000

rpm. The pellets were let to dry and then resuspended in Laemli 1x.

The cells were washed with PBS. Then a small amount of lysis buffer ( 50 mM Tris-HCl pH 7.5,

150 mM NaCl, 1% NP-40, 2 mM EDTA) was added to the cells. The cells were removed from

the wells and transferred to an Eppendorf . The samples were then incubated on ice for 30

minutes. Then they were centrifuged for 5 minutes at 13 000 rpm at 4C. After centrifugation, the

supernatant from each sample was collected in eppendorf tubes. The pellets were recuperated

56

and washed with -20C acetone and centrifugated again at 13 000 rpm. The pellets were let to dry

and then resuspended in Laemli 1x.

3.5.2 - Samples preparation

Then the protein sample was mixed to the Laemmli buffer in a ration 1 : 0,5. The volume was

then completed to 40 ul of H2O. The complete mix was then boiled for 5 minutes and then

cooled at room temperature for 5 minutes.

3.5.3 - Western Blot

Protein collected from cells culture were separated using gel electrophoresis on a 10%

polyacrylamide gel. The separated proteins were transferred onto nitrocellulose membranes and

blocked in 5 % Bovine Serum Albumin (BSA)-0.1% Phosphate Buffer Saline-Tween (PBS-T)

for an hour. The membranes were probed with anti-Noggin (ORIGENE co.- 1:2000 dilution with

5 % BSA-0.1% TBS-T) for an hour at room temperature. Subsequently, the membranes were

washed twice with 0.1% PBS-T and incubated with Horseradish peroxidase (HRP) conjugated

anti-Mouse IgG (1:25000 dilution in 1 % BSA-0.1% TBS-T) for an hour at room temperature.

Following incubation with the secondary antibody, the membranes were washed three times

using 0.1% TBS-T. Signal was detected by chemiluminescence after incubation of the blots with

the ECL+ reagent (Amersham) exposure to film. The same process was performed using anti-

Chordin (R and D systems),1:2000 dilution with 5 % BSA-0.1% TBS-T) but this time using an

HRP-conjugated anti-goat IgG 1: 25000.

57

3.6 - Alkaline phosphatase assay

Finally we wanted to see if our knockdown would be related to an increase osteogenic

differentiation. The alkaline phosphatase was then answering our question.

First the cell cultures medium was removed and then the cultures were rinsed with Tris Buffered

Saline (TBS) three times. Then for each well, lysis buffer 62.5 µl ( 20 mM Tris ph 10 + 0.5 mM

MgCl2 + 0.1% Triton X-100). Then a freeze-thaw (-20C) was performed to extract the ALP from

the cells. Then, 25 ul of the cells extracts was added to 75 ul of ALP reaction buffer (100 mM

Tris+ 100 mM NaCl, pH 9.5) and 100 ul of p-Nitrophenyl phosphate liquid substrate (SIGMA).

We incubated our samples at 37C. The activity of the alakaline phosphatase enzyme was

measured by the appearance of the p-Nitrophenol (pNP) product, which gives a yellow color,

every 5 mins for a period of 45 mins. Quantification of pNP was analyzed through a standard

curve. The protein content was quantified using the Bradford assay. The data are expressed as the

amount of pNP produced per min/ug of protein.

58

4 - MATERIALS & METHODS IN VIVO STUDIES

The second part of the project was to see if we would be able to transfer our findings from our in

vitro studies to an in vivo model. We decided to study the 2 most potent shRNAs, one targeting

Noggin and the second targeting Chordin and to use them in a wild-type mice model. Once

again, we would use them through a lentiviral vector to inject them at the distraction site for a

localized increase osteogenesis.

DO was performed on the right tibia of 54 wild-type mice using a miniature Illizarov distraction

device. Animals were randomized into two major groups according to the time of sacrifice: end

of distraction (day 17) and mid-consolidation (day 34). Each major group was sub-divided in 3

subgroups representing the type of injection that was administered at the site of distraction on

day 8. For each time point, 9 mice were injected with PBS, 9 with a lentivirus plasmid containing

a Non-Target (NT) shRNA and 9 with lentivirus plasmids containing shRNAs targeting Noggin-

Chordin combined together. To evaluate the success of the infection a GFP marker was added to

lentivirus plasmid targeting Noggin. For each subgroup 6 collected samples were studied using

Faxitron X-ray, µCT and immunohistochemistry and 3 were studied using Rt-PCR.

59

Day Procedure

0 Surgery day

5 Start distraction

8 Injection

11 End of distraction / 1st sacrifice

14 Mid-consolidation / 2nd sacrifice

Table 1 : Timeline animal procedures during in vivo studiesThis timeline explain the different steps and procedures that were effectuated on the animals during the in vivo studies

4.1 - Operative and distraction protocol

The surgical procedure was approved by the McGill University Animal Care Committee. DO

was performed on strain C57Bl/6 male mice, aged 2-3 months obtained from Charles River

using a miniature Ilizarov fixator adapted from Jill Helms study (Tay et al., 1998). Mice were

subcutaneously injected with 0.1 ml of (1mg/kg-Sigma) of buprenorphine and then anesthetized

with isoflurane using a Fortec pump (Cyprane Keighley, England) throughout surgery

(approximately 30 minutes). After shaving the right leg, two 0.25 mm insect pins (Austerlitz,

Marlborough, MA) were drilled 90° apart into the proximal metaphysis of the right tibia. Both

pins remained perpendicular to the tibia. The pins were secured into position using a ring and

four hexagonal nuts. A second set of pins was drilled into the distal metaphysis of the right tibia

approximately 12 mm away from the proximal set of pins. Three rods were inserted through the

60

first ring of the mini-Ilizarov fixator (Paolo Alto, CA) (Figure 7 and Figure 8) and secured into

position. A second ring was placed through the three rods and secured onto the distal set of pins

using hexagonal nuts. Once the mini-fixator was installed into place, a small incision was made

along the diaphysis of the right tibia using a no.11 surgical scalpel (Fisher Scientific, Osaka,

Japan). A small hole was drilled into the middle of the diaphysis using an insect pin to weaken

the bone and facilitate the osteotomy. Using the scalpel, the bone was cut along the hole to

complete the osteotomy. The fractured ends of the bone were aligned and the incision was closed

using coated vicryl sutures (J385H-Johnson & Johnson, North Ryde, NSW). Animals were

subcutaneously injected with 0.1 ml of buprenorphine and checked regularly throughout a 5 day

latency period for pain management before initiating distraction. Mice were allowed to weight

bear immediately. Mice were distracted for 12 days using a distraction rate of 0.2 mm every 12

hours. Mice were euthanized by using carbon dioxide. Distracted tibial samples were collected at

2 time points: 17 days (end of distraction phase) and 34 days (mid-consolidation phase) for

further analysis.

61

Figure 8: Mini Ilizarov external fixator

Small scale Ilizarov external fixator developped by Dr. Jill Helms in Stanford University, Palo Alto, CA

Figure 9 : Mouse in DO

Mouse with mini Ilizarov external fixator applied on a wild-type mice

62

4.2 - Injection

4.2.1 - Transfer of noggin shRNA into lentivirus plasmid containing GFP

The plasmid containing the active Noggin shRNA was changed to replace the puromycin cassette

with GFP. To do so, a plasmid containing a GFP coding sequence was digested with EcoRI and

the fragment purified on a minelute column. This fragment was then inserted into the Noggin

shRNA plasmid that was previously restricted with EcoRI, thus removing the puromycin

sequence. The resulting plasmid was confirmed by digestion with HinDIII.

N420 noggin shRNA lentiviral plasmid7397 bp

GFP

ampi

part of Env GP160 glycoprotein

TRCN0000066294 (N420)

extra C in more recent Sigma sequence5'GAG D3rdG

PsiSD

HIV 5'-LTR

pUCoriSV40pAU5'

RSIN/3'-LTR

partial U3PPT

RRE

pLKO-REVpLKO-FWD

primer REV GFP

primer FWD GFP

CMV

RSV

U6

bla promoter

Figure 10: Final plasmid containing GFP after Noggin shRNA transfer

Position of Noggin shRNA in plasmid containing GFP after transfer

63

4.2.2 - Injection protocol

For the injection 3 different groups were formed. The first group received an injection of shRNA

lentiviruses targetting Chordin and Noggin together , the second group received NT shRNAS

lentiviruses end the third group received PBS. The injection was realized as follow : On day 8

post-surgery, a 10 ul percutaneous bolus injection was realized using a 100 ul insulin syringe.

4.3 - Faxitron X-ray and Micro-Computed Tomography

In preparation for micro-computed tomography (µCT) and Faxitron x-ray, distracted bones

collected from wild-type and heterozygous mice (n=6 per time point) were fixed in 4%

paraformaldehyde (PFA) 4g of paraformaldehyde in 100 ml 1x PBS) overnight at 4°C. Samples

were washed in 1x PBS for 30 minutes and consecutively dehydrated in graded solutions of 50%

and 70% ethanol twice for one hour intervals at room temperature. Samples stored in 70%

ethanol were taken to the Centre of Bone and Periodontal Research of McGill University for

analysis.

µCT analysis was completed using the SkyScan 1072 (Aartselaar, Belgium) that has a 20-100

KeV/0-250 mA sealed, air-cooled, microfocus X-ray source with a polychromatic beam. The

SkyScan 1072 was also equipped with a detector that contained a 12-bit, cooled CCD camera

(1024 by 1024 pixels) coupled by a fiber optics taper to the scintillator. The distracted tibiae were

scanned at 45 KeV/222 µA with 25X magnification (11.25 µm pixel size). Image reconstruction

was performed using NRecon (1.4.4, SkyScan). Static histomorphometry parameters such as,

tissue volume (mm3), bone volume (mm3), bone volume/tissue volume (BV/TV %) were

64

measured using the CT Analyser (1.8.0.2, SkyScan). Furthermore, the Faxitron MX-20 was used

to produce radiographs of the distracted tibiae.

4.4 - Immunohistochemistry

4.4.1 - Paraffin embedding

Collected tibial samples from 17 days (end of distraction) and 34 days (mid-consolidation phase)

were suspended in 4% PFA overnight at 4°C. The following day, samples were washed in 1x

PBS for 30 minutes. Samples were then submerged in a solution of 0.85% saline/30% ethanol for

15 minutes. The samples underwent a gradual dehydration process by immersing them in

solutions of 50%, 70%, 90% and 100% ethanol for 15 minute each. The dehydrated tibial

samples were resuspended in solutions of 1:1 ethanol:xylene (Fisher Scientific) and xylene for

15 minutes each. Moreover, the samples were washed three times with molten paraplast (Oxford

Labware, St. Louis, MO) and then kept in paraplast overnight at 55°C under vacuum. The

samples were positioned in molten paraplast-containing cassettes and left to solidify at 4°C

forseveral hours. After paraffinization, the blocks were trimmed at 0.7 µm and sectioned

longitudinally at 5 µm using a Lecia RM 2255 microtome (Leica Microsystems, Richmond Hill,

ON). Sections were warmed in a 30°C water bath for 30 seconds and then placed on Superfrost

Plus slides (Scientific Device Laboratory, Des Plaines, IL).

65

4.4.2 - GFP marking

Distracted tibial samples (n=3 per time point) were deparaffinized and blocked with 10%

hydrogen peroxide for 10 minutes to inhibit endogenous peroxidase activity. The sections were

washed with distilled water and 1x PBS (phosphate buffered saline). Following the washings, the

samples were incubated in phosphate-buffered saline containing 10% normal goat serum (Vector

Labs, Burlingame, CA) in a humidified chamber for 20 minutes to block nonspecific binding.

Sections were rinsed with 1x PBS and blocked once more using the avidin-biotin blocking kit

(Vector, SP-2001). For immunostaining, commercially available GFP antibodies were used to

detect GFP marking in a dilution 1/50.

4.5 - RT-PCR on tibia sample

4.5.1 - Bone collection

First the tibia bone specimens were collected from the mice. As much soft tissue and skin were

removed. They the specimen was place in a 15 ml tube in an RNA later solution. Then the tube

was placed at -20oC for future steps, until all specimens are ready for analysis.

4.5.2 - Tissue Homogenisation

First the bone was place in a fresh Petri dish and only the distracted zone was cutted in small

pieces. The crushed tissue was suspended in 2 ml of Trizol (Invitrogen) and further homogenized

using a Polytron PT-MR 3000 (Kinematica) for 20 seconds at 20 000 rpm.

66

4.5.3 - RNA extraction, reverse transcription RNA and RT-PCR Noggin and Chordin

The RNA extraction, the reverse transcription of RNA and the RT-PCR for Noggin and Chordin

followed the same protocols as described earlier for the extract from the cells cultures.

4.5.4 - RT - PCR : GFP and puromycin

The PCR reactions were carried out in a final volume of 25 ul containing 2.5 µl of 10x PCR

Buffer- MgCl2 (200 mM Tris-HCl pH 8.4, 500 mM KCl, 1.25 ml MgCl2), 0.5 µl of 10 mM

dNTP, 0.5 µl of each oligonucleotides primers 25 uM (GFP and puromycin) , 0.25 µl of 5U/µl

Taq DNA polymerase (Invitrogen, Carlsbad, CA) and 0.1 ug of cDNA. The primer sequences

for GFP and puromycin are as followed : GFP-FWD (CTCGTGACCACCCTGACC) GFP-REV

(TCTGCTGGTAGTGGTCGG), PURO-FWD (GTACAAGCCCACGGTGCG) PURO-REV

(TCGGCGGTGACGGTGAAG). The PCR reactions were amplified using a GeneAMP PCR

System 9700 thermal cycler (Applied Biosystems, Singapore). PCR conditions included heating

for 5 minutes, 35 cycles of denaturation at 94°C for 30 seconds, annealing at 58°C for 30

seconds and extension at 72°C for 45 seconds, with further extension at 72°C for 7 minutes. The

expected size for puromycin and GFP fragments were 534 bp and 376 bp respectively. The PCR

products were separated via electrophoresis using a 1.3% UltraPure Agarose (Invitrogen) gel.

The PCR products were visualized by Ethidum Bromide staining.

67

5- RESULTS IN VITRO

5.1 - RT-PCR results non-transfected MC3T3-E1

MC3T3-E1 cells were seeded and grown to confluence (designated as day 0). Cells were then

differentiated in a-MEM with ascorbic acid and beta-glycerophosphate. Then we proceeded with

RT-PCR at different timepoints to be able to evaluate the time when the RNA expression for both

Noggin and Chordin was at its highest level. In both cases, the levels of RNA expression of

Noggin and Chordin was at its highest on Day 7.

Graph 1 : RT-PCR -MC3T3-E1 Noggin and Chordin mRNA level

MC3T3-E1 cells were seeded and grown to confluence (designated as day 0). Cells were then differentiated in a-MEM with ascorbic acid and beta-glycerophosphate. Then we proceeded with RT-PCR at different timepoints to be able to evaluate the time when the RNA expression for both Noggin and Chordin was at its highest level. In both cases, the levels of RNA expression of Noggin and Chordin was at its highest on Day 7.

68

5.2 - RT-PCR on transduced MC3T3-E1 cultures

Because we found that Day 7 was the day with the highest RNA expression for both Noggin and

Chordin, we decided to study our knockdown at that timepoint. After transduction of MC3T3-E1

cells, stably expressing pools were studied through qRT-PCR analysis showed that the shRNAs

were effective in knocking down Noggin endogenous mRNA levels down to 10% compared to

the control- NT.

Graph 2 : Noggin mRNA levels in stable pools MC3T3-E1 expressing individual shRNA through lentiviral infection

In MC3T3-E1 cells stably expressing shRNAs targeting Noggin, on day 7 post-confluence, qRT-PCR analysis showed that the shRNAs shRNA 1 was effective in knocking down Noggin endogenous mRNA levels down to 10%.

69

Also, after transduction of MC3T3-E1 cells, qRT-PCR analysis showed that the shRNAs were

effective in knocking down Chordin endogenous mRNA levels down to 17% compared to the

control-NT.

Graph 3 : Chordin mRNA levels in stable pools MC3T3-E1 expressing individual shRNA through lentiviral infection

In MC3T3-E1 cells stably expressing shRNAs targeting Noggin, on day 7 post-confluence, qRT-PCR analysis showed that the shRNAs 1 was effective in knocking down Chordin endogenous mRNA levels down to 17%.

Those results are consistent with a successful knockdown of Noggin and Chordin mRNA level in

MC3T3-E1 cells after our transduction.

70

5.3 - Western Blot

Culture of MC3T3-E1 stably expressing shRNAs targetting Noggin were grown and the proteins

from the media and the cells were collected and studied through Western Blot analysis. The

results showed a successful knockdown of the Noggin protein by shRNA 1 for both the proteins

secreted in the condition media and the proteins from the cells. Only the 2 most successful

shRNA have been shown in the following images.

Figure 11 : Noggin - Western blot on secreted proteins in condition media of MC3T3-E1 stably expressing shRNAs targeting Noggin1-Noggin recombinant proteins, 2-NT, 3-shRNA 2, 4-shRNA 1Culture of MC3T3-E1 stably expressing shRNAs targeting Noggin were grown and the proteins from the media and the cells were collected and studied through Western Blot analysis. The results showed a successful knockdown of the Noggin protein by shRNA 1 for the proteins secreted in the condition media

71

Figure 12 : Noggin - Western blot on cellular proteins of MC3T3-E1 stably expressing shRNAs targeting Noggin 1-Noggin recombinant proteins, 2-NT, 3-shRNA 2, 4-shRNA 1Culture of MC3T3-E1 stably expressing shRNAs targeting Noggin were grown and the proteins from the media and the cells were collected and studied through Western Blot analysis. The results showed a successful knockdown of the Noggin protein by shRNA 1 for the cellular proteins

The same study was performed for the shRNA targetting Chordin on an MC3T3-E1 culture

stably expressing shRNAs targeting Chordin. The results showed a successful knockdown of

Chordin in the proteins secreted in the condition media. Unfortunately our experiment was not

able to provide clear results that could be interpreted for the cellular proteins.

72

Figure 13 : Chordin Western blot secreted proteins in condition media of MC3T3-E1 stably expressing shRNAs targeting Chordin1- Noggin recombinant proteins, 2-NT, 3-shRNA 2, 4-shRNA 1Culture of MC3T3-E1 stably expressing shRNAs targeting Chordin were grown and the proteins from the media and the cells were collected and studied through Western Blot analysis. The results showed a successful knockdown of the Chordin protein by shRNA 1 for the proteins secreted in the condition media

73

5.4 - Phosphatase alkaline assay

The phosphatase alkaline activity is a marker of the osteogenic differentiation. The test was

performed on cell cultures stably expressing the most potent shRNA from our previous test (RT-

PCR and Western Blot) for Noggin and Chordin. The ALP assays were done on day 2 and day 4

post confluence. They were normalized via the Bradford Assay.

They were compared to the control NT.

The results showed an increase of the phosphatase alkaline activity that was significant for

Noggin at both time points. For Chordin the results also showed an increase activity on day 2,

the difference compared to the control (NT) was not significant on day 4.

74

Graph 4 : ALP activity in MC3T3-E1 stably expressing the shRNA targeting Noggin and ChordinThe test was performed on cell cultures stably expressing the most potent shRNA from our previous test (RT-PCR and Western Blot) for Noggin and Chordin. The ALP assays were done on day 2 and day 4 post confluence. They were normalized via the Bradford Assay.The alkaline phosphatase activity is significantly increase in cells stably expressing shRNAs targeting Noggin and Chordin on Day 2.

75

6 - IN VIVO RESULTS

6.1 - Surgical and distraction protocol success rate

Distraction osteogenesis was performed on a total of 56 2-3 months old wild-type mice. Of the

56 mice, 54 successfully survived DO, accounting for a 96.4 % success rate post-surgery. The 2

mice died post-surgery due to the development of a necrotic foot.

6.2 - Injection success rate

An injection of either PBS, lentiviral plasmids including NT shRNA or lentiviral plasmids

including shRNAs targetting Noggin and Chordin was done on day 8 post surgery.

Unfortunately, at the time of the injection a significant volume of the injection was coming out of

injection site. This complication was observed for most of the injections realized.

6.3 - Faxitron x-ray

Distracted tibial samples were collected from wild-type mice at 17 and 34 days post-surgery

( end of distraction and mid-consolidation). For each group (Noggin-Chordin, NT, PBS) 6

samples were studied for each timepoints. Bone fill scores were utilized to evaluate the

consolidation rate between the distracted segments.

Three independent and blinded reviewers evaluated the Faxitron x-ray results of mice belonging

to different dosing groups sacrificed at end of distraction (day 17) and mid-consolidation (day

34).

76

75

Chapter 4.0-Results

4.1-Mid-Consolidation Phase

4.1.1-Faxitron x-ray

Distracted tibial samples were collected from C57Bl/6 wild type male mice at 34 and 51 days

post-surgery (mid-consolidation and end of consolidation respectively) (Figure 6). Bone fill

scores were utilized to evaluate the consolidation rate between the distracted segments (Table 1).

Table 1. Bone-fill scores definition

Bone-Fill Scores Radiographic Evaluation

0 No visible bone-fill (0%)

1 Visible bone-fill, but less than 50%

2 More than 50% visible bone-fill, but less than 100% visible bone-fill

3 100% visible bone-fill

Three independent and blinded reviewers evaluated the Faxitron x-ray results of mice belonging

to different dosing groups sacrificed at mid-consolidation (day 34) and end of consolidation

(51d).

Table : Bone filling scoresSubjective Bone-Fill scores assign to the Faxitron images obtained

Unfortunately, there were no visible bone-filling score difference that could be noted in between

the treatment group and the two control groups at both timepoints.

77

Graph 5 : Bone filling scoresBone filing scores were given for each of the samples. After analysis of the results, no difference could be noted in the bone filling scores between the treatment group Noggin-Chordin and the 2 controls

78

6.4 - Micro-Computed Tomography (µ-CT)

Distracted tibial samples were collected from the wild type mice at 17 and 34 days

corresponding to end of distraction and mid-consolidation respectively. Samples (6 for each

group at each timepoints), a total of 36, were analyzed using µ-CT.

6.4.1 - Bone volume

Bone volume was calculated by the same blinded technician at McGill Bone Center.

Unfortunately for all three groups, at both timepoints no statistical difference could be measure

in terms of bone volume.

79

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

Noggin/Chordin NT PBS

BV

(mm

^3)

Treatment group

Bone Volume (BV)

17 days

34 days

Graph 6 : Bone volumesBone volume was measured using the uCT images for the samples (n=6) of each groups. After analysis, no statistically significant difference could be noted in between the Noggin/Chordin group and the two controls.

80

6.4.2 - Bone volume/Tissue volume ratio

The same results could be observe for the calculation of the ration of Bone Volume / Tissue

volume. No statistically significant difference could be observe between the treatment 3 groups.

0.00

5.00

10.00

15.00

20.00

25.00

30.00

Noggin/Chordin NT PBS

BV/

TV

Treatment group

Bone Volume/Tissue Volume (BV/TV)

17 days

34 days

Graph 7 : Bone volume / Tissue volume ratiosThe ratio of Bone volume/ Tissue volume was measured using the uCT images for the samples (n=6) of each group. After analysis, no statistically significant difference could be noted in between the Noggin/Chordin group and the two controls.

81

6.5 - RT-PCR

RT-PCR studies were realized on the distracted zone of the tibia for all the groups. At each

timepoints a for each group 3 samples were collected. After analysis, at both 17 and 34 days, the

results did not show any significant knockdown of Noggin or Chordin.

Also, there were no detectable and specific amplification products for both and GFP and

Puromycin.

Chordin mRNA levels in stable pools MC3T3-E1 expressing individual shRNA through lentiviral infection

Graph 8 : RT-PCR tibia samplesRT-PCR were realized on distracted tibia samples (n=3) for each group. No statistically significant knockdown of Noggin and Chordin could be observed in the treatment group compared to the controls at both timepoints (17 and 34 days).

82

6.6 - Immunohistochemistry - GFP marking

For the in vivo studies, the lentiviral plasmid containing the shRNA targeting Noggin, the

puromycin segment was changed for GFP segment. As we were proceeding with our

immunohistochemistry studies, we were expecting to found signal in the samples that had

received the injection of lentiviruses for Noggin and Chordin. However, as the experiment

started, to be able to find any GFP marking we had to greatly increase the sensitivity of our test.

When we finally obtained results, we found GFP marking in all 3 groups : Noggin-Chordin, NT

and PBS. Most probably we had increased the sensitivity of our test so much to get detection that

we had lost the specificity of our test.

83

Puromycin and GFP signal in MC3T3-E1 cells stably expressing shRNAs targeting Noggin and Chordin

Figure 14 : Puromycin and GFP signal in MC3T3-E1 cells stably expressing shRNAs targeting Noggin and ChordinThere were no detectable and specific amplification products for both and GFP and Puromycin in the Noggin and Chordin group.

84

7- DISCUSSION

Multiple technique and treatment have been investigated to accelerate bone formation during

DO. The use of BMPs have been investigated in the past and seems to be presenting promising

results and participate to an accelerated bone formation in DO [108].

However, one of the main concerns with BMPs are the large concentrations of these proteins

needed to be administered at the distraction site with unknown side effects [109] and high costs.

Thus, an alternative method is manipulation of the intrinsic pathways to accelerate bone

formation during DO. This could be achieved by manipulating the endogenous BMP pathways in

order to up regulate their expression. So our hypothesis was that by inhibiting the BMP

antagonists Noggin and Chordin we would increase osteogenesis. So far no study had been made

of knockdown of Noggin and Chordin in an in vivo model of DO.

For this reason, we first tested our hypothesis in an in vitro setting. A knockdown of Noggin and

Chordin had been tried using a shRNAs targeting both of them though a lentiviral plasmid

delivery system. The cells culture chosen was the MC3T3-E1 because of their preosteoblastic

nature. First our RT-PCR studies confirmed that we achieved an efficient knockdown of the

mRNA levels for both Chordin and Noggin by the most potents shRNAs. Following those

results, the decision to evaluate if this knockdown would translate in a protein knockdown.

Western blot analysis were made for both Noggin and Chordin. A successful knockdown of

Noggin for both cellular proteins and culture media proteins was found. For Chordin, also a

successful knockdown was proven for the cellular proteins. The next step was then to see if those

85

successful knockdown would translate in an increase osteogenic activity. The ALP assay showed

an increased activity for Noggin at both 2 and 4 days post-confluence and for Chordin at day 2.

Encouraged by those results we decided to transport our study to an in vivo model. We decided

to use our most efficient shRNAs targetting Noggin and Chordin (one for each) and use them in

our in vivo studies. We used an animal model of wild-type mice that underwent distraction

osteogenesis using a small scale Ilizarov external fixator. The treatment group was receiving an

injection on day 8 post-surgery with lentiviruses containing each 1 shRNA targeting Noggin or

Chordin. The control groups were receiving either an injection of lentiviruses containing a Non-

Target (NT) shRNA or a PBS injection. I operated on a total of 56 mice and 54 survived the

surgery and post-operative period. Unfortunately, during the experimentation our main issue was

related to the injection. Most of the injected solution was extravasating from the injection site. It

was also very difficult to assess the volume of injection loss because the liquid formula was

getting loss on the animal skin/hair.

Faxitron and uCT were realized to evaluation the osteogenesis at both time points (17 and 34

days). A total of 6 samples for each group at each timepoints were sent to the bone center for

analysis. Sadly, none of the results showed a significant increase in osteogenesis in the treatment

group compared to the controls. Also, our RT-PCR study (performed on 3 samples/group/

timepoint) was concordant with those results as it did not show any significant knockdown of

Noggin and Chordin in the treatment group compared to the controls. Finally, it is interesting to

see that GFP marking was found in all 3 groups while we were only expecting it for the Noggin-

86

Chordin group. Also, no GFP RNA could be identify in our PCR study performed on the

distracted tibia samples from the treatment group Noggin and Chordin. At the light of those

results, we decided to stop our immunohistochemistry studies.

At this point, I think the main issue with our results are related to the injection technique. The

proper way of injection, the volume of injection and the consistency of the injection should be

eventually reevaluated. But it is certain that if most of the injection does not reach the site that

we wish to treat, it is very difficult to imagine that we would get any significant difference in

between the groups. Possible solutions would be to use a systemic delivery system that would

target specifically to the bone-formation surfaces. Zhang et al. were recently able to develop a

targeted delivery system for RNA interference-based anabolic therapy using (AspSerSer)6-

liposome. [110]

However, our disappointing in vivo results could also be related to other physiologic cascades

that could be activated by our knockdown. It is easy to think that other BMPs antagonists could

be upregulated in response to a knockdown of Noggin and Chordin. This would then open the

hypothesis that knockdown of Noggin and Chordin might not be enough to achieve an increased

osteogenesis. Although this idea goes in contradiction with previous successful studies

performed for example by Wan and Al [78] where they obtained an accelerated bone formation

with Noggin knockdown.

Finally it is important to think that this in vivo study was mainly constructed as a proof of

concept since there are still concerns related to the usage of lentiviral delivery system.

Theorically, because thee site of integration is unpredictable, the provirus can disturb the

87

function of cellular genes and lead to activation of oncogenes promoting the development of

cancer.However, studies have shown that lentivirus vectors have a lower tendency to integrate in

places that potentially cause cancer than gamma-retroviral vectors.[111] More specifically, one

study found that lentiviral vectors did not cause either an increase in tumor incidence or an

earlier onset of tumors in a mouse strain with a much higher incidence of tumors.[112]

This is why, the next step would be to find another efficient delivery system. Different options

are available to us, but so far the nanoparticles delivery system has been the one that have

interested us the most.

In conclusion, we were able to show a successful knockdown of Noggin and Chordin in an in

vitro model. Our ALP assay was also consistent with and increased osteogenic differentiation

activity. Our in vivo study was interesting by being one of the first one to study Noggin and

Chordin knockdown in a DO animal model. Unfortunately, our results were not able to confirm

either a successful knockdown or an increased osteogenesis.

88

8-References

1. Ilizarov, G.A. and A.A. Deviatov, [Surgical elongation of the leg]. Ortop Travmatol Protez,

1971. 32(8): p. 20-5.

2. Ilizarov G. The tension-stress effect on the genesis and growth of tissues: Part II. The

influence of the rate and frequency of distraction. Clinical orthopaedics and related research.

1989;239: 263-85.

3. Urist, M.R., Bone: formation by autoinduction. Science, 1965. 150(698): p. 893-9.

4. Ilizarov G. Operative Principles of Ilizarov. Italy: A.S.A.M.I. Group; 1990. 3-8.

5. Ducy P, Karsenty G. Genetic control of cell differentiation in the skeleton. Current Opinion

in Cell Biology. 1998;10(5): 614-19.

6. Vaes BLT, Ducy P, Sijbers AM, Hendriks JMA, van Someren EP, de Jong NG, et al.

Microarray analysis on Runx2-deficient mouse embryos reveals novel Runx2 functions and

target genes during intramembranous and endochondral bone formation. Bone. 2006;39(4):

724-38.

7. Tsiridis E, Upadhyay N, Giannoudis P. Molecular aspects of fracture healing: Which are the

important molecules? Injury. 2007;38(1S): 11-25.

8. Kosher RA. Collagen gene expression during limb cartilage differentiation. The Journal of

Cell Biology. 1986;102(4): 1151-56.

89

9. Provot S, Schipani E. Molecular mechanisms of endochondral bone development.

Biochemical and Biophysical Research Communications. 2005;328(3): 658-65.

10. Hartmann C. Skeletal Development-Wnts Are in Control. Molecules and Cells. 2007;24(2):

177.

11. Kojimoto H, Yasui N, Goto T, Matsuda S, Shimomura Y. Bone Lengthening In Rabbits By

Callus Distraction. Journal of Bone & Joint Surgery, British Volume. 1988(70-B): 543-9.

12. Delloye C, Delefortrie G, Coutelier L, Vincent A. Bone regenerate formation in cortical bone

during distraction lengthening. An experimental study. Clin Orthop Relat Res. 1990;250:

34-42.

13. Arnonson J, Harrison BH, Stewart CL, Harp Jr JH. The histology of distraction osteogenesis

using different external fixators. Clin Orthop Relat Res. 1989;241: 106-16.

14. Rauch F, Lauzier D, Croteau S, Travers R, Glorieux FH, Hamdy R. Temporal and spatial

expression of bone morphogenetic protein-2,-4, and-7 during distraction osteogenesis in

rabbits. Bone. 2000;27(3): 453-59.

15. Yasui N, Sato M, Ochi T, Kimura T, Kawahata H, Kitamura Y, et al. Three Modes of

Ossification During Distraction Osteogenesis in The Rat. Journal of Bone & Joint Surgery,

British Volume. 1997;79(5): 824-30.

16. Yasui N, Sato M, Ochi T, Kimura T, Kawahata H, Kitamura Y, et al. Three Modes of

Ossification During Distraction Osteogenesis in The Rat. Journal of Bone & Joint Surgery,

British Volume. 1997;79(5): 824-30.

17. Paley D. Problems, obstacles, and complications of limb lengthening by the Ilizarov

technique. Clin Orthop Relat Res. 1990;250: 81-104.

90

18. Greenwald JA, Luchs JS, Mehrara BJ, Spector JA, Mackool RJ, McCarthy JG, et al.

Pumping the regenerate: An evaluation of oscillating distraction osteogenesis in the rodent

mandible. Annals of plastic surgery. 2000;44(5): 516-21.

19. Kim UK, Chung IK, Lee KH, Swift JQ, Seong WJ, Ko CC. Bone Regeneration in

Mandibular Distraction Osteogenesis Combined With Compression Stimulation. Journal of

Oral and Maxillofacial Surgery. 2006;64(10): 1498-505.

20. Malizos KN, Hantes ME, Protopappas V, Papachristos A. Low-intensity pulsed ultrasound

for bone healing: An overview. Injury. 2006;37(1S): 56- 62.

21. Sakurakichi K, Tsuchiya H, Uehara K, Yamashiro T, Tomita K, Azuma Y. Effects of timing of

low-intensity pulsed ultrasound on distraction osteogenesis. Journal of Orthopaedic

Research. 2004;22(2): 395-403.

22. Eberson CP, Hogan KA, Moore DC, Ehrlich MG. Effect of low-intensity ultrasound

stimulation on consolidation of the regenerate zone in a rat model of distraction osteogenesis.

Journal of pediatric orthopedics. 2003;23(1): 46-51.

23. Shimazaki A, Inui K, Azuma Y, Nishimura N, Yamano Y. Low-intensity pulsed ultrasound

accelerates bone maturation in distraction osteogenesis in rabbits. Journal of Bone & Joint

Surgery, British Volume. 2000;82(7): 1077-82.

24. Claes L, Rüter A, Mayr E. Low-intensity ultrasound enhances maturation of callus after

segmental transport. Clinical orthopaedics and related research. 2005(430): 189-94.

25. Kitoh H, Kitakoji T, Tsuchiya H, Mitsuyama H, Nakamura H, Katoh M, et al.

Transplantation of marrow-derived mesenchymal stem cells and platelet- rich plasma during

distraction osteogenesis—a preliminary result of three cases. Bone. 2004;35(4): 892-98.

91

26. Tsubota S, Tsuchiya H, Shinokawa Y, Tomita K, Minato H. Transplantation of osteoblast-like

cells to the distracted callus in rabbits. Journal of Bone & Joint Surgery, British Volume.

1999;81(1): 125-29.

27. Takamine Y, Tsuchiya H, Kitakoji T, Kurita K, Ono Y, Ohshima Y, et al. Distraction

osteogenesis enhanced by osteoblastlike cells and collagen gel. Clinical orthopaedics and

related research. 2002(399): 240-46.

28. Riggs BL, Parfitt AM. Drugs used to treat osteoporosis: the critical need for a uniform

nomenclature based on their action on bone remodeling. J Bone Miner Res. 2005;20(2):

177-84.

29. Takahashi M, Yukata K, Matsui Y, Abbaspour A, Takata S, Yasui N. Bisphosphonate

modulates morphological and mechanical properties in distraction osteogenesis through

inhibition of bone resorption. Bone. 2006;39(3): 573-81.

30. Yeung H, Lee S, Fung K, Leung K. Expression of basic fibroblast growth factor during

distraction osteogenesis. Clin Orthop Relat Res. 2001;385: 219-29.

31. Greenwald JA, Luchs JS, Mehrara BJ, Spector JA, Mackool RJ, McCarthy JG, et al.

Pumping the regenerate: An evaluation of oscillating distraction osteogenesis in the rodent

mandible. Annals of plastic surgery. 2000;44(5): 516-21.

32. Kitoh H, Kitakoji T, Tsuchiya H, Katoh M, Ishiguro N. Transplantation of culture expanded

bone marrow cells and platelet rich plasma in distraction osteogenesis of the long bones.

Bone. 2007;40(2): 522-28.

33. Tiedeman, J.J., et al., Treatment of nonunion by percutaneous injection of bone marrow and

demineralized bone matrix. An experimental study in dogs. Clin Orthop Relat Res,

1991(268): p. 294-302.

92

34. Oikarinen, J., Experimental spinal fusion with decalcified bone matrix and deep-frozen

allogeneic bone in rabbits. Clin Orthop Relat Res, 1982(162): p. 210-8.

35. Glowacki, J., D. Altobelli, and J.B. Mulliken, Fate of mineralized and demineralized osseous

implants in cranial defects. Calcif Tissue Int, 1981. 33(1): p. 71-6.

36. Fowler, E.B., L.G. Breault, and G. Rebitski, Ridge preservation utilizing an acellular dermal

allograft and demineralized freeze-dried bone allograft: Part II. Immediate endosseous

implant placement. J Periodontol, 2000. 71(8): p. 1360-4.

37. Bolander, M.E., Balian G. The use of demineralized bone matrix in the repair of segmental

defects. Augmentation with extracted matrix proteins and a comparison with autologous

grafts.J Bone Joint Surg Am,1986. 68(8) : p. 1264-74

38. Einhorn, T.A., et al., The healing of segmental bone defects induced by demineralized bone

matrix. A radiographic and biomechanical study. J Bone Joint Surg Am, 1984. 66(2): p.

274-9.

39. Gepstein, R., R.E. Weiss, and T. Hallel, Bridging large defects in bone by demineralized bone

matrix in the form of a powder. A radiographic, histological, and radioisotope- uptake study

in rats. J Bone Joint Surg Am, 1987. 69(7): p. 984-92.

40. Hagino, T. and Y. Hamada, Accelerating bone formation and earlier healing after using

demineralized bone matrix for limb lengthening in rabbits. J Orthop Res, 1999. 17(2): p.

232-7.

41. Giannoudis PV, Dinopoulos H, Tsiridis E, Bone substitutes: an update. Injury 2005, 36(Suppl

3):S20-27.

42. Finkemeier CG, Bone-grafting and bone-graft substitutes.J Bone Joint Surg Am 2002, 84(3):

454-464.

93

43. Bullens PH, Bart Schreuder HW, de Waal Malefijt MC, Verdonschot N, Buma P: Is an

impacted morselized graft in a cage an alternative for reconstructing segmental diaphyseal

defects? Clin Orthop Relat Res 2009, 467(3):783-791.

44. Ostermann PA, Haase N, Rübberdt A, Wich M, Ekkernkamp A: Management of a long

segmental defect at the proximal meta-diaphyseal junction of the tibia using a cylindrical

titanium mesh cage. J Orthop Trauma 2002, 16(8):597-601.

45. Wang, Y., Ni M., Tang PF., Li G.,Novel application of HA-TCP biomaterials in distraction

osteogenesis shortened the lengthening time and promoted bone consolidation., J Orthop

Res. 2009 Apr;27(4):477-82

46. Akkouch A, Zhang Z, Rouabhia M: A novel collagen/hydroxyapatite/poly(lactide-co-ε-

caprolactone) biodegradable and bioactive 3D porous scaffold for bone regeneration. J

Biomed Mater Res A 2011, 96A:693-704.

47. Tampieri A, Landi E, Valentini F, Sandri M, D'Alessandro T, Dediu V, Marcacci M: A

conceptually new type of bio-hybrid scaffold for bone regeneration. Nanotechnology 2011,

22(1):015104.

48. Laschke MW, Witt K, Pohlemann T, Menger MD: Injectable nanocrystalline hydroxyapatite

paste for bone substitution: in vivo analysis of biocompatibility and vascularization.

J Biomed Mater Res B Appl Biomater 2007, 82(2):494-505.

49. Bail, H.J., et al., Systemic application of growth hormone for enhancement of secondary and

intramembranous fracture healing. Horm Res, 2002. 58 Suppl 3: p. 39-42.

50. Byung-Hyun Park, S.J.Y., Kyu Yun Jang, Mi-Ran Kim, Hyung-Seok Lee, Ki-Bum Kim, and

S.Y.L. Hyuk Park, Ho Sung Park, Seok Tae Lim, Kyung-Jin Song, Jung Ryul Kim, COMP–

94

angiopoietin-1 accelerates bone formation during distraction osteogenesis. Bone, 2010. 46: p.

6.

51. Xiao, Y.T., L.X. Xiang, and J.Z. Shao, Bone morphogenetic protein. Biochem Biophys Res

Commun, 2007. 362(3): p. 550-3.

52. Issack, P.S. and P.E. DiCesare, Recent advances toward the clinical application of bone

morphogenetic proteins in bone and cartilage repair. Am J Orthop, 2003. 32(9): p. 429- 36.

53. Yoon,B.S. and K.M. Lyons, Multiple functions of BMPs in chondrogenesis. J Cell Biochem,

2004. 93(1): p. 93-103.

54. Luyten, F.P., and al., Purification and partial amino acid sequence of osteogenin, a protein

initiating bone differentiation. J Biol Chem, 1989. 264(23): p. 13377-80.

55. Attisano, L., and al., Identification of human activin and TGF beta type I receptors that form

heteromeric kinase complexes with type II receptors. Cell, 1993. 75(4): p. 671-80.

56. Riggs, B.L. and A.M. Parfitt, Drugs used to treat osteoporosis: the critical need for a uniform

nomenclature based on their action on bone remodeling. Journal of Bone & Mineral

Research, 2005. 20(2): p. 177-84.

57. Heldin, C.H., K. Miyazono, and P. ten Dijke, TGF-beta signalling from cell membrane to

nucleus through SMAD proteins. Nature, 1997. 390(6659): p. 465-71.

58. Kawabata, M., T. Imamura, and K. Miyazono, Signal transduction by bone morphogenetic

proteins. Cytokine Growth Factor Rev, 1998. 9(1): p. 49-61.

59. Urist, M.R., Bone: formation by autoinduction. Science, 1965. 150(698): p. 893-9.

95

60. Mizumoto Y, Moseley T, Drews M, CooperIii VN, Reddi AH. Acceleration of Regenerate

Ossification During Distraction Osteogenesis with Recombinant Human Bone

Morphogenetic Protein-7. Journal of Bone & Joint Surgery. 2003;85(90003): 124-30.

61. Yang JH, Kim HJ, Kim SE, Yun YP, Bae JH, Kim SJ, Choi KH, Song HR,The effect of bone

morphogenic protein-2-coated tri-calcium phosphate/hydroxyapatite on new bone formation

in a rat model of femoral distraction osteogenesis, Cytotherapy. 2012 Mar;14(3):315-26.

Epub 2011 Nov 28.

62. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, Miyazono K, Smurf1

interacts with transforming growth factor-beta type I receptor through Smad7 and induces

receptor degradation, J. Biol. Chem. 276 (2001) 12477–12480.

63. Garrison KR, Shemilt I, Donell S, Ryder JJ, Mugford M, Harvey I, Song F, Alt V Bone

morphogenetic protein (BMP) for fracture healing in adults. Cochrane Database Syst Rev.

2010 Jun 16;(6):CD006950.

64. Ziad Dahabreh, Rozalia Dimitriou, Peter Giannoudis: Health economics: A cost analysis of

treatment of persistent fracture non-unions using bone morphogenetic protein-7. Injury 38,

371-377 (2007)

65. Elliot Carlisle, Jeffrey Fischgrund: Bone morphogenetic proteins for spinal fusion. Spine J

5(6 Suppl), 240S-249S (2005)

66. Thomas Kleeman, Uri Michael Ahn, Anne Talbot- Kleeman: A Laparoscopic anterior lumbar

interbody fusion with rhBMP-2. Spine 26(24), 2751-2756 (2001)

67. Zara JN, Siu RK, Zhang X, Shen J, Ngo R, Lee M, Li W, Chiang M, Chung J, Kwak J, Wu

BM, Ting K, Soo C.High doses of bone morphogenetic protein 2 induce structurally

abnormal bone and inflammation in vivo. Tissue Eng Part A. 2011 May;17(9-10):1389-99.

Epub 2011 Mar 3.

96

68. Garrison KR, Donell S, Ryder J, Shemilt I, Mugford M, Harvey I, Song F. Clinical

effectiveness and cost-effectiveness of bone morphogenetic proteins in the non-healing of

fractures and spinal fusion: a systematic review. Health Technol Assess. 2007 Aug;11(30):

1-150, iii-iv.

69. Dean BJ, Watson T, Moed BR, Zhang Z, Role of bone morphogenetic proteins and their

antagonists in healing of bone fracture, Frontiers in Bioscience 14 2009 p. 2878-2888,

70. Rosen V., BMP and BMP inhibitors in bone. Ann N Y Acad Sci, 2006. 1068: p. 19-25.

71. Brown, M.A., et al., Crystal structure of BMP-9 and functional interactions with pro-region

and receptors. J Biol Chem, 2005. 280(26): p. 25111-8.

72. Hillger, F., et al., Biophysical comparison of BMP-2, ProBMP-2, and the free pro- peptide

reveals stabilization of the pro-peptide by the mature growth factor. J Biol Chem, 2005.

280(15): p. 14974-80.

73. Aspenberg P, Charlotte Jeppsson, Aris Economide: The bone morphogenetic proteins

antagonist noggin inhibits membranous ossification. J Bone Miner Res 2001 16(3), 497-500

74. Gazzerro E, Canalis E: Bone morphogenetic proteins and their antagonists. Rev Endo Meta

Disorders 2006 7(1-2),51-65

75. Yanagita M: BMP antagonists: their roles in development and involvement in

pathophysiology. Cytokine and Growth Factor Re 2005 16, 309-317

76. Wijerde M, Karp S, McMahon J, McMahon A: Noggin antagonism of BMP4 signaling

controls development of the axial skeleton in the mouse. Dev Biol 2005 286, 149-157

77. Gazzerro E, Gangji V, Canalis E: Bone morphogenetic proteins induce the expression of

noggin, which limits their activity in cultured rat osteoblasts. J Clin Invest 1998 102(12),

2106–2114

97

78. Wan D, Pomerantz J, Brunet L, Kim JB, Chou YF, Wu B, Harland R, Blau H, Longaker M:

Noggin suppression enhances in Vitro osteogenesis and accelerates in Vivo bone formation. J

Bio Chem 2007 282(36), 26450-26459

79. Niikura T, Hak D, Reddi AH: Global gene profiling reveals a downregulation of BMP gene

expression in experimental atrophic nonunions compared to standard healing fractures. J

Ortho Res 2006 24(7), 1463-1471

80. Piccolo S, Sasai Y, Lu B, De Robertis E: Dorsoventral patterning in Xenopus: inhibition of

ventral signals by direct binding of chordin to BMP-4. Cell 1996 86, 589 -598.

81. Zhang D, Ferguson CM, O’Keefe R, Puzas JE, Rosier R, Reynolds P: A role for the BMP

antagonist chordin in endochondral ossification. J Bone Miner Res 2002 17(2), 293-300

82. Balemans W, Van Hul W: Extracellular regulation of BMP signaling in vertebrates: A

cocktail of modulators. Dev Biol 2002 250, 231-250

83. Iemura S, Yamamoto T, Takagi C, Uchiyama H, Natsume T, Shimasaki S, Sugino H, Ueno N:

Direct binding of follistatin to a complex of bone-morphogenetic protein and its receptor

inhibits ventral and epidermal cell fates in early Xenopus embryo. Proc Natl Acad Sci USA

1998 95, 9337-9342

84. Nilsson O, Parker E, Hegde A, Cha M, Barnes K, Baron J: Gradients in bone morphogenetic

protein-related gene expression across the growth plate. J Endocrinology 2007 193, 75-84

85. Pushparaj PN, Aarthi JJ, Manikandan J, Kumar SD, siRNA, miRNA, and shRNA: in vivo

Applications J Dent Res 87(11) 2008 p.992-1003

86. Rao DD, Vorhies JS, Senzer N, Nemunaitis J. siRNA vs. shRNA: similarities and

differences.Adv Drug Deliv Rev. 2009 Jul 25;61(9):746-59. Epub 2009 Apr 20.

87. Cullen BR, RNAi the natural way, Nat. Genet. 37 (2005) 1163–1165.

98

88. Lee Y, C. Han AJ, Choi H, Kim J, Yim, J. Lee J, Provost P, Radmark O, Kim S, Kim VN,

The nuclear RNase III Drosha initiates microRNA processing, Nature 425 (2003) 415–419.

89. Zhang H, Kolb FA, Brondani V, Billy E, Filipowicz W, Human Dicer preferentially cleaves

dsRNAs at their termini without a requirement for ATP, EMBO J. 21 (2002) 5875–5885.

90. Lee Y, Jeon K, Lee JT, Kim S, Kim VN, MicroRNA maturation: stepwise processing and

subcellular localization, EMBO J. 21 (2002) 4663–4670.

91. Cullen BR, Transcription and processing of human microRNA precursors, Mol. Cell 16

(2004) 861–865.

92. Yi R, Qin Y, Macara IG, Cullen BR, Exportin-5 mediates the nuclear export of pre-

microRNAs and short hairpin RNAs, Genes Dev. 17 (2003) 3011–3016.

93. Lee YS, Nakahara K, Pham JW, Kim K, He Z, Sontheimer EJ, Carthew RW, Distinct roles

for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways, Cell 117

(2004) 69–81.

94. Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U, Nuclear export of microRNA

precursors, Science 303 (2004) 95–98.

95. Landthaler M, Gaidatzis D, Rothballer A, Chen PY, Soll SJ, Dinic L, Ojo T, Hafner M,

Zavolan M, Tuschl T, Molecular characterization of human Argonaute-containing

ribonucleoprotein complexes and their bound target mRNAs, RNA 14 (2008) 2580–2596.

96. Check E, A tragic setback, Nature 420 (2002) 116–118.

97. Nguyen T, Menocal EM, Harborth J, Fruehauf JH, RNAi therapeutics: an update on delivery,

Curr. Opin. Mol. Ther. 10 (2008) 158–167.

99

98. Luten J, Van Nostrum CF, De Smedt SC, Hennink WE, Biodegradable polymers as non-viral

carriers for plasmid DNA delivery, J. Control. Release 126 (2008) 97–110.

99. Zhang S, Zhao B, Jiang H, Wang B, Ma B, Cationic lipids and polymers mediated vectors for

delivery of siRNA, J. Control. Release 123 (2007) 1–10.

100. Aigner A, Nonviral in vivo delivery of therapeutic small interfering RNAs, Curr. Opin. Mol.

Ther. 9 (2007) 345–352.

101. Naldini L et al. In vivo delivery and stable transduction of nondividing cells by a lentiviral

vector. Science 1996; 272: 263-267.

102. An DS, Xie Y, Mao SH, Morizono K, Kung SK, Chen IS, Lentiviral vectors for short

hairpin RNA delivery into human cells. Hum Gene Ther. 2003 Aug p.1207-12.

103. Kafri T., Blomer U., Peterson D.A., Gage F.H.,Verma I.M. Sustained expression of genes

delivered di- rectly into liver and muscle by lentiviral vectors. Nat. Genet. (1997) 17, 314–

317.

104. Miller DG, Adam MA, Miller AD. Gene transfer by retrovirus vectors occurs only in cells

that are actively replicating at the time of infection. Mol Cell Biol 1990; 10: 4239-4242.

105. Connolly JB, Lentiviruses in gene therapy clinical research, Gene Therapy (2002) 9, 1730–

1734

106. ShRNA Lentiviral Particles Transduction, Santa Cruz protocol

107. Bilezikian John P., Gideon Raisz L. Gideon A. R., Principles of bone biology : volume 1

(2002)

108. Rengachary, S.S., Bone morphogenetic proteins: basic concepts. Neurosurg Focus (2002) p.

e2.

100

109. Haque, T., et al., Characterizing the BMP pathway in a wild type mouse model of

distraction osteogenesis. Bone (2008), 1144-53.

110. Zhang G.; Guo B; Wu H.; Tang T. et al., A delivery system targeting bone formation

surfaces to facilitate RNAi-based anabolic therapy, Nature (2012) 18 (2) : 307-315

111. Cattoglio, C.; Facchini, G.; Sartori, D.; Antonelli, A.; Miccio, A.; Cassani, B.; Schmidt, M.;

Von Kalle, C. et al., Hot spots of retroviral integration in human CD34+ hematopoietic cells,

Blood (2007) 110 (6): 1770–1778

112. Montini, E.; Cesana, D.; Schmidt, M.; Sanvito, F.; Ponzoni, M.; Bartholomae, C.; Sergi, L.

S.; Benedicenti, F. et al., Hematopoietic stem cell gene transfer in a tumor-prone mouse

model uncovers low genotoxicity of lentiviral vector integration, Nature Biotechnology

(2006) 24 (6): 687–696

101