162
Interactions between Undifferentiated and Osteogenically Differentiated Mesenchymal Stromal Cells during Osteogenesis Yinghong Zhou BDS, MDS Institute of Health and Biomedical Innovation School of Chemistry, Physics and Mechanical Engineering (CPME) Faculty of Science & Engineering Queensland University of Technology Thesis submitted for the award of Doctor of Philosophy February 2013

Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

Interactions between Undifferentiated and

Osteogenically Differentiated Mesenchymal

Stromal Cells during Osteogenesis

Yinghong Zhou BDS, MDS

Institute of Health and Biomedical Innovation

School of Chemistry, Physics and Mechanical Engineering (CPME)

Faculty of Science & Engineering

Queensland University of Technology

Thesis submitted for the award of Doctor of Philosophy

February 2013

Page 2: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted
Page 3: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

i | P a g e

KEYWORDS

Bone regeneration, bone tissue engineering, cell-based therapy, cell interaction, cell

recruitment, mesenchymal stromal cells (MSCs), osteogenesis, osteogenic

differentiation, vascular endothelial growth factor (VEGF), CXCL12/CXCR4,

phosphoinositide 3-kinase (PI3K) /Akt

Page 4: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

ii | P a g e

ABSTRACT

Despite tremendous efforts that have been made to develop new therapeutic strategies,

including the fabrication of novel bioactive scaffolds, the application of various

growth factors and gene-modified cells in pre-clinical or clinical settings, bone defect

healing still poses a great challenge to orthopaedic surgeons.

Increasing evidence supports that cell-based therapy has emerged as one of the most

promising therapeutic approaches for tissue repair and regeneration due to the

inherent characteristics of mesenchymal stromal cells (MSCs) in respect to their self-

renewal capacity and multipotent differentiation potential. More interestingly, recent

experimental evidence suggests that implanted MSCs have a close communication

with host cells, and the contribution of donor cells is beyond their direct conversion

into bone forming cells. Collectively, these findings indicate that transplanted donor

cells play an important role in bone defect healing. However, how transplanted donor

cells initiate osteogenesis, their role in the recruitment of host MSCs to form

functional structures and the mechanisms behind these processes remain largely

undefined. Motivated by the issues mentioned above, the purpose of this thesis was to

unveil the interactions between donor cells and host cells during osteogenesis and in

particular the involvement of VEGF, the CXCL12/CXCR4 axis and the PI3K/Akt

signaling pathway in cell recruitment and donor-host cell interactions. This project

was designed as three separate but closely-linked segments which, in the end, led to

three independent papers.

The first segment of this project investigated how osteogenically differentiated human

mesenchymal stromal cells (O-hMSCs) were involved in the osteogenic processes and

how they interacted with the host cells after implantation. Histology,

immunohistochemistry and in situ hybridisation were some of the techniques applied

Page 5: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

iii | P a g e

in this part of the study to identify the interaction between donor cells and host cells in

vivo. Results revealed that in both ectopic (subcutaneous pocket) and orthotopic (skull

defect) sites of severe combined immunodeficiency (SCID) mice, donor O-hMSCs

could recruit host cells to initiate osteogenesis. We demonstrated that the ratio of

donor cells and host cells inside the implants was significantly dependent on the local

micro-environment and directly correlated with osteogenesis. The findings from this

study provided clear evidence that donor O-hMSCs play an integral role in host cell

recruitment during osteogenesis.

Osteogenesis and angiogenesis are two closely correlated processes during functional

bone formation and regeneration. As the best characterised angiogenic mediator,

VEGF is believed to play a crucial role in skeletal development by enhancement of

the coupling of angiogenesis and osteogenesis. The second segment of this project

was designed to investigate the role of VEGF in the interaction between

undifferentiated and osteogenically differentiated bone marrow stromal cells (MSCs

and OMSCs) in vitro and in vivo. VEGF secretion increased dramatically in MSCs

after osteogenic differentiation. Furthermore, the secretion of VEGF from OMSCs

altered the expression pattern of CXCL12/CXCR4 in MSCs in vitro, which may be

responsible for the increased cell migration in vivo. In situ hybridisation demonstrated

that donor OMSCs recruited host cells during the osteogenic processes at the

orthotopic sites, and blocking VEGF with neutralizing antibody resulted in a

significant decrease of host cell recruitment and new bone formation. These findings

suggest that the interaction between donor OMSCs and host MSCs might be mediated

by the paracrine effect of VEGF and the activation of CXCL12/CXCR4 axis,

resulting in enhanced host MSCs migration to the implantation site.

Based on the findings of the second segment, subsequent experiments were designed

to determine the possible mechanisms that drive the host cells to migrate to the defect

site. Our co-culture studies demonstrated that OMSCs induced phenotypic changes in

MSCs via dimerization of platelet-derived growth factor receptor (PDGFR), which

then activated the signal transduction. The PI3K/Akt signaling pathway was probed

Page 6: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

iv | P a g e

by using targeted PI3K inhibitor LY294002, and the activity was monitored by

Western blotting analysis using total- and phospho-Akt specific antibodies.

Downstream targets of the PI3K/Akt signaling pathway, mTOR, p70S6k

and PAK1,

which regulate actin organization and cell motility, were also activated in the presence

of OMSCs-derived conditioned medium. The findings from this segment

demonstrated that VEGF secreted by OMSCs plays a critical role in recruiting host

cells to accelerate osteogenesis via the activation of PDGFRs, which then activate the

PI3K/Akt signaling pathway.

In summary, the body of work presented in this dissertation has demonstrated that the

interactions between donor cells and host cells were critical for the initiation of

ectopic and orthotopic osteogenesis and that this process was mediated by the VEGF,

CXCL12/CXCR4 and the PI3K/Akt signaling pathway. The findings from this

dissertation may provide a scientific rationale for the development of novel

therapeutic strategies in the treatment and management of bone defects.

Page 7: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

v | P a g e

TABLE OF CONTENTS

KEYWORDS ............................................................................................................. i

ABSTRACT .............................................................................................................. ii

TABLE OF CONTENTS .......................................................................................... v

LIST OF FIGURES ................................................................................................. vii

LIST OF ABBREVIATIONS ................................................................................... ix

STATEMENT OF ORIGINAL AUTHORSHIP ...................................................... xii

LIST OF PUBLICATIONS .................................................................................... xiii

LIST OF CONFERENCE PRESENTATIONS ........................................................ xv

ACKNOWLEDGEMENTS ................................................................................... xvii

CHAPTER 1 INTRODUCTION................................................................................. 1

1.1 Preface ........................................................................................................ 1

1.2 Main purpose of this study .......................................................................... 2

1.3 Questions to be addressed............................................................................ 2

1.4 Possible outcomes and significance ............................................................. 3

CHAPTER 2 OVERVIEW OF PROJECT DESIGN .................................................... 4

2.1 The effect of pre-programmed donor cell on host cell recruitment during

osteogenesis ........................................................................................................... 4

2.2 Possible regulatory factors that lead to the recruitment of host cells ............. 5

2.3 Potential cell signaling pathways involved in the host cell recruitment ........ 6

CHAPTER 3 LITERATURE REVIEW ....................................................................... 7

3.1 Overview .................................................................................................... 7

3.2 Cell-based therapy for bone defects ............................................................. 8

3.3 Cell sources for cell-based bone tissue engineering ................................... 10

Page 8: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

vi | P a g e

3.4 The contribution of donor cells in cell-based bone tissue engineering ........ 15

3.5 The involvement of host cells in cell-based bone tissue engineering .......... 16

3.6 Chemokines and growth factors related with cell migration ....................... 19

3.7 Signaling pathways related with cell migration .......................................... 27

3.8 Summary ................................................................................................... 34

CHAPTER 4 RESEARCH REPORTS ...................................................................... 35

REPORT ONE..................................................................................................... 37

Implantation of osteogenically differentiated donor mesenchymal stromal cells

causes recruitment of host cells ............................................................................ 37

REPORT TWO .................................................................................................... 55

VEGF induced CXCL12/CXCR4 axis in the recruitment of mesenchymal stromal

cells (MSCs) during osteogenesis......................................................................... 55

REPORT THREE ................................................................................................ 81

Involvement of VEGF/PDGFR and the PI3K/Akt signaling pathway in OMSCs-

induced MSCs migration ..................................................................................... 81

CHAPTER 5 CONCLUSIONS AND PERSPECTIVES ............................................103

5.1 General discussion ..................................................................................104

5.2 Project summary .....................................................................................113

5.3 Future direction .......................................................................................115

REFERENCES.......................................................................................................117

Page 9: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

vii | P a g e

LIST OF FIGURES

Figure 1. Schematic of cell-based therapy strategy .................................................... 9

Figure 2. Schematic of osteoblast differentiation..................................................... 13

Figure 3. Schematic of endogenous cells homing to the damaged tissues for in situ

tissue regeneration ................................................................................................... 17

Figure 4. CXCL12/CXCR4 signaling pathway ....................................................... 22

Figure 5. Vascular endothelial growth factor (VEGF) family members and their

receptors .................................................................................................................. 24

Figure 6. Simplified schematic of the platelet-derived growth factor (PDGF) system

................................................................................................................................ 26

Figure 7. Signaling through phosphatidylinositol (3,4,5)-triphosphate .................... 29

Figure 8. Schematic of the three mammalian mitogen-activated protein kinase

(MAPK) signaling pathways ................................................................................... 33

Figure 9. hMSCs characterisation and in vitro osteogenic differentiation ................ 45

Figure 10. In vitro migration assay. ........................................................................ 46

Figure 11. Osteogenesis in subcutaneous and skull defect implants ......................... 48

Figure 12. Distribution of donor human cells and host mouse cells in the

subcutaneous implants revealed by ISH ................................................................... 49

Figure 13. Distribution of O-hMSCs and host mouse cells in the skull defect implants

and bone marrows, revealed by ISH ........................................................................ 51

Figure 14. Histological evaluation of a skull defect model after a 4-week implantation

of OMSCs ............................................................................................................... 68

Page 10: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

viii | P a g e

Figure 15. VEGF secretion from OMSCs ............................................................... 69

Figure 16. VEGF neutralizing antibody interfered with the migration of MSCs

towards OMSCs. ..................................................................................................... 71

Figure 17. Morphological and cytoskeletal changes in MSCs cultured in CMOMSC

with or without the addition of VEGF neutralizing antibody .................................... 72

Figure 18. CMOMSC

increased the CXCL12/CXCR4 expression of MSCs ............... 73

Figure 19. CXCL12/CXCR4 expression of MSCs stimulated by VEGF.................. 74

Figure 20. Blocking VEGF with neutralizing antibody resulted in a significant

decrease of MSCs recruitment and new bone formation ........................................... 76

Figure 21. Schematic of the proposed mechanism by which donor OMSCs initiate a

paracrine signaling cascade that results in altered phenotype and enhanced motility of

host MSCs in a skull defect model ........................................................................... 79

Figure 22. The expression of VEGFR and PDGFR mRNA transcripts was examined

by RT-qPCR analysis, using HUVECs as positive control ....................................... 90

Figure 23. CMOMSC

-induced tyrosine phosphorylation of PDGFR-α and PDGFR-β in

MSC. ....................................................................................................................... 92

Figure 24. The levels of protein expression of PDGFR-α and PDGFR-β in MSCs in

response to different stimulations ............................................................................ 93

Figure 25. The involvement of PDGFR in MSCs migration induced by CMOMSC

.... 94

Figure 26. Akt and Erk1/2 phosphorylation status in MSCs in response to various

stimulations ............................................................................................................. 96

Figure 27. Activation of the PI3K/Akt signaling is required for CMOMSC

induced

MSCs migration ...................................................................................................... 97

Figure 28 The phosphorylation status of downstream effectors of the PI3K/Akt

signaling pathway. ................................................................................................... 98

Page 11: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

ix | P a g e

LIST OF ABBREVIATIONS

ALP alkaline phosphatase

BM bone marrow

BMP bone morphogenetic protein

BMSC bone marrow stromal cell

BSA bovine serum albumin

BSP bone sialoprotein

COLI Type-I collagen

CXCL12 chemokine (C-X-C motif) ligand 12

CXCR4 chemokine (C-X-C motif) receptor 4

DAB 3, 3'-diaminobenzidine

DMEM Dulbecco's Modified Eagle Medium

DNA deoxyribonucleic acid

EC endothelial cell

ECM extracellular matrix

EDTA ethylenediaminetetraacetic acid

EGM endothelial cell growth media

EPC endothelial progenitor cell

ESC embryonic stem cell

FACS fluorescence activated cell sorting

Page 12: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

x | P a g e

FBS fetal bovine serum

HRP horseradish peroxidase

HUVEC human umbilical vein endothelial cell

IHC immunohistochemistry

ISH in situ hybridisation

kDa kilodalton

MSC mesenchymal stromal cell

OB osteoblast

OCN osteocalcin

O(-h)MSC osteogenically differentiated (human) mesenchymal

stromal cell

OPN osteopontin

PBS phosphate buffered saline

PDGF platelet-derived growth factor

PDGFR platelet-derived growth factor receptor

PI3K phosphoinositide 3-kinase

P/S penicillin and streptomycin

RNA ribonucleic acid

RNase ribonuclease

RT-qPCR real time quantitative reverse transcription polymerase

chain reaction

RUNX2 Runt-related transcription factor 2

SCID severe combined immunodeficiency

Page 13: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xi | P a g e

SDS-PAGE sodium dodecyl sulfate polyacrylamide gel

electrophoresis

TBS Tris-buffered saline

VEGF vascular endothelial growth factor

VEGFR vascular endothelial growth factor receptor

Page 14: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted
Page 15: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xiii | P a g e

LIST OF PUBLICATIONS

The following is a list of published, accepted or submitted manuscripts that have

arisen from the work in this thesis or are relevant to the work performed in this

PhD project:

1. Zhou Y, Fan W, Prasadam I, Crawford R, Xiao Y. Implantation of osteogenic

differentiated donor mesenchymal stem cells causes recruitment of host cells. Journal

of Tissue Engineering and Regenerative Medicine, 2012. doi: 10.1002/term.1619.

(IF: 3.278)

2. Wu C, Zhou Y, Fan W, Han P, Chang J, Yuen J, Zhang M, Xiao Y. Hypoxia-

mimicking mesoporous bioactive glass scaffolds with controllable cobalt ion release

for bone tissue engineering. Biomaterials, 2012. 33(7):2076-85. (IF: 7.404)

3. Zhou Y, Chakravorty N, Xiao Y, Gu W. Mesenchymal stem cells and nano-

structure surfaces. Book chapter in Methods in Molecular Biology, 2013. Springer.

Accepted.

4. Zhou Y, Crawford R, Xiao Y. VEGF induced CXCL12/CXCR4 axis in the

recruitment of mesenchymal stromal cells (MSCs) by osteogenically differentiated

MSCs (OMSCs) during osteogenesis. Manuscript submitted.

5. Zhou Y, Crawford R, Xiao Y. Involvement of VEGF/PDGFR and PI3K/Akt

signaling pathway in OMSCs induced MSCs migration. Manuscript submitted.

The following is a list of publications that are not relevant to the work performed

in this PhD project but published during the PhD candidature:

1. Wu C, Zhou Y (co-first author), Xu M, Han P, Chen L, Chang J, Xiao Y.

Copper-containing mesoporous bioactive glass scaffolds with multifunctional

Page 16: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xiv | P a g e

properties of angiogenesis capacity, osteostimulation and antibacterial activity.

Biomaterials, 2012. 34(2):422-33. (IF: 7.404)

2. Wu C, Zhou Y (co-first author), Lin C, Chang J, Xiao Y. Strontium-containing

mesoporous bioactive glass scaffolds with improved osteogenic/cementogenic

differentiation of periodontal ligament cells for periodontal tissue engineering. Acta

Biomaterialia, 2012. 8(10):3805-15. (IF: 4.865)

3. Zhou Y, Wu C, Xiao Y. The stimulation of proliferation and differentiation of

periodontal ligament cells by the ionic products from Ca7Si2P2O16 bioceramics. Acta

Biomaterialia, 2012. 8(6):2307-2316. (IF: 4.865)

4. Wu C, Fan W, Zhou Y, Luo Y, Gelinsky M, Chang J, Xiao Y. 3D-Printing of

highly uniform β-CaSiO3 ceramic scaffolds: preparation, characterization and in vivo

osteogenesis. Journal of Materials Chemistry, 2012. 22:12288-12295. (IF: 5.099)

5. Fan W, Wu C, Han P, Zhou Y, Xiao Y. Porous Ca-Si-based nanospheres: a

potential intra-canal disinfectant-carrier for infected canal treatment. Materials

Letters, 2012. 81:16-19. (IF: 2.307)

6. Wu C, Zhang Y, Zhou Y, Fan W, Xiao Y. A comparative study of mesoporous

glass/silk and non-mesoporous glass/silk scaffolds: Physiochemistry and in vivo

osteogenesis. Acta Biomaterialia, 2011. 7(5):2229-2236. (IF: 4.865)

7. Zhang Y, Fan W, Nothdurft L, Wu C, Zhou Y, Crawford R, Xiao Y. In vitro

and in vivo evaluation of adenovirus combined silk fibroin scaffolds for bone

morphogenetic protein-7 gene delivery. Tissue Engineering Part C: Methods, 2011.

17(8):789-797. (IF: 4.636)

8. Wu C, Zhang Y, Fan W, Ke X, Hu X, Zhou Y, Xiao Y. CaSiO3 microstructure

modulating the in vitro and in vivo bioactivity of poly (lactide-co-glycolide)

microspheres. Journal of Biomedical Materials Research: Part A, 2011. 98(1):122-

131. (IF: 3.044)

Page 17: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xv | P a g e

LIST OF CONFERENCE PRESENTATIONS

The following is a list of conference presentations during the PhD candidature:

1. Zhou Y, Fan W, Prasadam I, Han P, Zhang X, Crawford R, Xiao Y.

Recruitment of host mesenchymal stromal cells by osteogenic differentiated donor

osteoblasts via VEGF regulated pathway. IHBI Inspires Postgraduate Student

Conference. 22nd

-23rd

November 2012, Gold Coast, Australia. Oral presentation.

2. Zhou Y, Fan W, Prasadam I, Crawford R, Xiao Y. VEGF induced

CXCL12/CXCR4 axis in the recruitment of mesenchymal stem cells (MSCs) by

osteogenic differentiated bone marrow stromal cells (O-BMSCs). 3rd

Tissue

Engineering & Regenerative Medicine International Society (TERMIS) World

Congress 2012. 5th-8

th September 2012, Vienna, Austria. Oral presentation.

3. Zhou Y, Crawford R, Xiao Y. The migration of host cells towards osteogenic

differentiated donor mesenchymal stromal cells in ectopic and orthotopic

osteogenesis. 1st Asia-Pacific Bone and Mineral Research Meeting with the

Australian & New Zealand Bone & Mineral Society (ANZBMS) 22nd

Annual

Scientific Meeting. 2nd

-5th

September, 2012, Perth, Australia. Poster presentation.

4. Zhou Y, Crawford R, Xiao Y. Implantation of osteogenic differentiated donor

mesenchymal stromal cells causes recruitment of host cells. Australian & New

Zealand Orthopaedic Research Society (ANZORS) 18th

Annual Scientific

Meeting. 30th

August-1st September 2012, Perth, Australia. Oral presentation.

5. Zhou Y, Fan W, Crawford R, Xiao Y. Hypoxia maintains stemness of BMSCs,

PDLCs and DPCs. IHBI Inspires Postgraduate Student Conference. 24th-25

th

November 2011, Brisbane, Australia. Oral presentation.

Page 18: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xvi | P a g e

6. Zhou Y, Fan W, Crawford R, Xiao Y. Stem cell reprogramming genes are

differentially expressed in BMSCs, PDLCs and DPCs in response to hypoxic

environment. Australian & New Zealand Orthopaedic Research Society

(ANZORS) 17th

Annual Scientific Meeting. 1st-2

nd September 2011, Brisbane,

Australia. Oral presentation.

7. Zhou Y, Fan W, Zhang Y, Crawford R, Xiao Y. Localisation of human bone

marrow-derived osteoblasts in both ectopic and orthotopic osteogenic process. IHBI

Inspires Postgraduate Student Conference. 25th-26

th November 2010, Gold Coast,

Australia. Poster presentation.

8. Zhang Y, Fan W, Nothdurft L, Wu C, Zhou Y, Crawford R, Xiao Y. In vitro

and in vivo evaluation of adenovirus combined silk fibroin scaffolds for bone

morphogenetic protein-7 gene delivery. Tissue Engineering & Regenerative

Medicine International Society (TERMIS) 2010 Asia Pacific Meeting. 15th-17

th

September 2010, Sydney, Australia. Poster presentation.

9. Zhou Y, Fan W, Zhang Y, Crawford R, Xiao Y. Localisation of human bone

marrow-derived osteoblasts in both ectopic and orthotopic osteogenic process. Tissue

Engineering & Regenerative Medicine International Society (TERMIS) 2010

Asia Pacific Meeting. 15th-17

th September 2010, Sydney, Australia. Poster

presentation.

Page 19: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xvii | P a g e

ACKNOWLEDGEMENTS

At the end of my PhD candidature, I would like to extend my appreciation and

gratitude to the following individuals, for without them, this thesis would simply be

impossible.

First and foremost, I would like to express my sincere gratitude to my principal

supervisor, Professor Yin Xiao, for his invaluable guidance, innovative ideas and

continuous support throughout my PhD journey. I really cherish and appreciate his

motivation, enthusiasm and his willingness to extend support not only in my study but

also in my life. I am grateful to my associate supervisor, Professor Ross Crawford, for

his expertise and stimulating clinical skills. His optimism and encouragement has

inspired me to tackle the challenges during the candidature. I consider myself

extremely fortunate to have been granted such a great opportunity to work with both

of them.

My appreciation also goes to all the former and current colleagues in the Bone Group,

especially Dr Chengtie Wu, Dr Wei Fan, Dr Indira Prasadam, Ms Wei Shi, Ms

Pingping Han and Ms Xufang Zhang. Learning and working with them has always

been so enjoyable. The friendship established during my PhD journey has been very

important to me.

I would also like to show my gratitude to Dr Michael Doran, Dr Travis Klein, Dr

Ying Dong, Dr Roland Steck, Dr Siamak Saifzadeh, and other staff at the Institute of

Health and Biomedical Innovation and the Medical Engineering Research Facility.

Thanks for the great help and precious suggestions to my study.

Page 20: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

xviii | P a g e

A special note of appreciation goes to IHBI supporting staff, especially Mr David

Smith, Mr Dod Roshanbin, Mr Robert Smeaton, and Mr Scott Tucker, who keep our

lab running smoothly and well-organized.

I would also like to take this opportunity to thank my wonderful friends, especially Dr

Xuan Hu and Ms Ellen Fong. Their valued friendship and support has been a

motivation in completion of this project.

A heartfelt gratitude goes to Matthew Tuch for his generous patience and

encouragement in these important years of my life. Thanks for believing in me and

being there for me with a heart warming smile.

At last, I wish to dedicate my thesis to my parents for their forever and unconditional

care and love. They have worked hard and spared no effort to provide the best for me

to thrive.

Page 21: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

1 | P a g e

CHAPTER 1 INTRODUCTION

1.1 Preface

Orthopaedic surgeons have long held the aspiration to heal bone defects resulting

from traumatic insult, oncological resection, congenital deformities, and progressive

degenerative diseases. To realize this ambition, innumerable studies have been

conducted to advance autogeneic, allogeneic and even xenogeneic bone graft

therapies [1-3]. Although some favourable results have been achieved in animal

studies or clinical trials [4-6], the outcomes are far from satisfactory due to limited

availability, donor site morbidity, insufficient integration and immunological

repulsive reactions in the recipient against allo- or xenogeneic bone grafts [7].

Advances in materials science, cell and molecular biology, especially in the

knowledge of the underlying mechanisms of bone healing have thrown a new light on

the repair and regeneration of bone defects [8].

Cell-based therapy for bone regeneration offers a paradigm shift that may provide

alternative solutions from the traditional invasive surgeries. Significant efforts have

been devoted to characterizing the potential cell sources that are relatively abundant

and easily accessible. However, there has not been a universal understanding as to

which expansion conditions are optimal for the manufacture of MSCs intended for

bone repair and regeneration. For example, different cell seeding density [9-11],

vascular endothelial growth factor (VEGF) application [12-15] and low oxygen

tension [16-18] have all been reported to improve the longevity and the osteogenic

potency of MSCs. The induction of MSCs towards osteoblast-like cells prior to the

implantation is also found to favour more bone regeneration [19-21]. In previous

studies the concept of cell-based bone tissue engineering has been tested using

collagen type I matrices seeded with cells of osteogenic potential and implanted into

sites with osseous damage [22, 23]. New bone formation was found in defect sites

implanted with bone marrow derived-osteoblasts (OBs) at 4 weeks, while no obvious

Page 22: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

2 | P a g e

new bone formation was identified at the same time point without OBs transplantation

[23]. Therefore, focus has been directed on elucidating the intrinsic controls that keep

the transplanted donor cells alive and direct them along particular differentiation

pathways, which may interact with the microenvironment where the host

mesenchymal stromal cells (MSCs) reside [24]. MSCs exist in highly specialized

microenvironments in which their maintenance, proliferation, migration and

differentiation involve a complex interplay of many local and systematic signals

between MSCs and adjacent cells [25]. Even in simplified in vitro model systems, the

molecular and microenvironmental cues which are necessary to induce bone cell

differentiation have not yet been fully identified. Furthermore, concurrent studies

have suggested that an activation and chemotactic migration of host MSCs adjacent to

the defect sites is favourable for bone regeneration [26]. However, the interaction

between donor cells and host cells, and the recruitment mechanisms of host MSCs to

osseous defects are not clear and need to be further demonstrated. In addition, the

differentiation state of the donor cells, which may have significant implications for

the effectiveness of cell transplantation, has not been sufficiently elucidated.

1.2 Main purpose of this study

The main purpose of this study is to investigate the interactions between donor cells

and host cells, and to identify the possible molecular mechanisms in the initiation of

bone formation upon donor cell implantation, which is essential to developing optimal

strategies for clinical application of cell-based therapy in bone tissue engineering.

1.3 Questions to be addressed

1. Do transplanted donor cells play a role in recruiting host MSCs to the

bone defect sites to initiate osteogenesis? How do donor cells interact

with host MSCs?

2. What are the possible regulatory factors from osteogenically

differentiated MSCs (OMSCs) that lead to the recruitment of host

MSCs?

Page 23: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

3 | P a g e

3. What are the potential signaling pathways involved in the

communication between donor cells and host cells during osteogenesis?

1.4 Possible outcomes and significance

Based on earlier attempts to deliver osteoprogenitor cells that are capable of

responding to the local microenvironment and facilitate new bone formation in a

robust and rapid fashion, the concept and application of cell-based therapy for bone

tissue engineering has been emphasized in this project. More knowledge about the

role of pre-programmed donor cells in bone defects healing, the interaction between

donor cells and host cells during osteogenesis has been revealed. Furthermore, this

project is believed to be the first to unveil the possible molecular pathways which may

induce the host cells’ response to the signals from the implanted exogenous cells, and

activate the bone repair and regeneration process upon cell implantation. The new

findings from this project will be of significant use in facilitating the application of

pre-programmed donor cells and hopefully extend the field of clinical applications of

cell-based therapy in bone tissue engineering.

Page 24: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

4 | P a g e

CHAPTER 2 OVERVIEW OF PROJECT DESIGN

2.1 The effect of pre-programmed donor cells on host cell

recruitment during osteogenesis

2.1.1 In vitro cell migration

In order to investigate the effect of manipulation of donor cells on the motility of

MSCs, an in vitro migration assay was performed using transwell system. Transwell

inserts seeded with MSCs were assembled onto the companion plates, which were

seeded with osteogenically differentiated MSCs (OMSCs) or undifferentiated MSCs.

After 8 h of incubation, cell motility was quantified by counting the number of cells

that migrated through the pores in response to different stimulations from the bottom

companion plates, and the differences were compared between groups.

2.1.2 In vivo host cell recruitment

Collagen scaffolds carrying OMSCs or MSCs were implanted subcutaneously or into

skull defects in severe combined immunodeficiency (SCID) mice. After samples were

harvested and fixed, micro-computed tomography (μCT) scanning was performed to

reconstruct three-dimensional (3D) images of the implants, and to measure the

calcification volume for statistical analysis. After samples were processed and

sectioned, the ectopic and orthotopic osteogenesis was assessed by subsequent

histological and immunohistochemical (IHC) staining. In situ hybridisation (ISH)

against human Alu sequences was performed to distinguish donor human cells from

host mouse cells, and to investigate the recruitment tendency of host cells in different

microenvironments. The localisation of transplanted human cells identified by the Alu

probe in both subcutaneous and skull defect scaffolds was observed, recorded and

analysed.

Page 25: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

5 | P a g e

2.2 Possible regulatory factors that lead to the recruitment

of host cells

2.2.1 Comparison between OMSCs and MSCs

In order to unveil the secreted factors from donor OMSCs that may lead to the

migration of host MSCs, further study in the new bone forming area was performed.

Triple staining technique revealed that donor OMSCs strongly expressed VEGF,

which might exert a paracrine effect resulting in host cell recruitment and their

involvement in new bone formation. This finding formed the foundation for the

subsequent project development. Alizarin Red S staining was performed to compare

the mineralization, while real time quantitative-PCR (RT-qPCR) analysis, enzyme-

linked immunosorbent assay (ELISA) and Western blotting analysis were performed

to compare the VEGF secretion between OMSCs and MSCs.

2.2.2 The in vitro regulatory effect of VEGF

Indirect co-culture models were applied to study the effect of VEGF derived from

OMSCs on the motility, the cell morphology and the CXCL12/CXCR4 expression of

MSCs. In vitro migration assay and real-time xCELLigence system were performed to

investigate the motility of MSCs in response to different stimulations, and the

responding cell morphology changes were further examined by fluorescent staining.

Furthermore, the expression pattern of CXCL12/CXCR4, which is the best known

axis that contributes to cell migration, was studied extensively and the effect of VEGF

on cell responses was probed using a specific neutralizing antibody.

2.2.3 The in vivo regulatory effect of VEGF

The regulatory effect of VEGF on host cell recruitment was examined by the

application of neutralizing VEGF antibody in vivo, after OMSCs implantation into the

skull defects in SCID mice. Histological staining and µCT scanning were performed

to observe the new bone formation. The recruitment of host cells to the bone defect

sites was monitored by ISH staining.

Page 26: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

6 | P a g e

2.3 Potential cell signaling pathways involved in the host

cell recruitment

2.3.1 Functional receptors profile

MSCs were stimulated with different concentrations of VEGF, or conditioned

medium derived from MSCs (CMMSC

) or OMSCs (CMOMSC

) to establish the receptors’

activation profile. The functional role of platelet-derived growth factor receptors

(PDGFRs) was further investigated using phospho-receptor tyrosine kinase (RTK)

array, Western blotting analysis and in vitro migration assay.

2.3.2 The involvement of cell signaling pathways

In order to unveil the potential regulatory cell signaling pathway involved in the host

cell recruitment, MSCs were stimulated in different conditions with or without the

interference of the dimerization of PDGFRs. The involvement of PI3K/Akt and

Erk1/2 in OMSCs-induced MSCs migration was monitored by relevant

phosphorylated antibodies and inhibitors. The time course phosphorylation status of

the PI3K/Akt signaling pathway was further investigated by Western blotting analysis.

2.3.3 Downstream effectors of the PI3K/Akt signaling pathway

The stimulatory effect of VEGF secreted from OMSCs on the activation of the

PI3K/Akt signaling pathway was further investigated by detecting the

phosphorylation status of the downstream effectors, mTOR, p70S6K

, and PAK1, by

Western blotting evaluation of specific antibodies.

Page 27: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

7 | P a g e

CHAPTER 3 LITERATURE REVIEW

3.1 Overview

Tissue engineering is an emerging interdisciplinary field involving principles of the

life sciences and engineering with the expectation of constructing a biological

substitute for damaged tissues, both in vitro and in vivo, in order to restore, maintain,

or improve normal tissue structure and function [27]. Rapid expansion of knowledge

regarding the potential for tissue engineering, and more specifically progenitor cell-

based tissue engineering, offers new insights to the treatment of an array of diseases,

such as graft-versus-host disease (GVHD) [28], cardiovascular failure [29, 30], liver

diseases [31], injury or degeneration of the spinal cord [32, 33], tendon [34], cartilage

[35], bone [36] and numerous others. Among the aforementioned conditions and

diseases, bone defects resulting from trauma or pathological and physiological

resorption represent a significant global health issue that deserves optimal treatment.

Although many of the conventional modalities for repairing bone defects are able to

restore certain levels of function, various drawbacks ranging from limited donor tissue

to procedure-related complications remain a major clinical challenge [7, 37]. Unlike

traditional surgeries that need to sacrifice tissues from the donor site, the tissue

engineering approach can achieve the goal of bone repair and regeneration without the

necessity of donor site morbidity [38]. Recent studies have demonstrated the

feasibility of generating tissue-engineered bone for repairing bone defects [39-41],

and the outcomes of ongoing clinical trials are very promising [36, 42, 43].

The concept of cell-based tissue engineering has fuelled investigations of various

progenitor cell sources that can be manipulated, either in vitro or in vivo, to repair or

replace damaged tissues. Given the complex three-dimensional structure of bone

tissues, cells, extracellular matrix, cell-cell communications, cell-matrix interactions,

growth factors, cytokines and other cell components have to be combined in a well-

coordinated space- and time-dependent manner to achieve successful results in bone

Page 28: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

8 | P a g e

tissue engineering. In the past few years, there have been significant advances in the

understanding of the molecular mechanisms of cell fate determination, an extremely

complex process regulated by multi-mutual interplay of signaling pathways [44-47].

Therefore, cell-based therapies with potential to stimulate endogenous progenitor cells

during osteogenesis have become a prime translational goal.

This review focuses on the current status of knowledge as well as the emerging

challenges in the area of cell-based therapy and potential cell sources related to bone

tissue engineering. Additionally, the critical role of the transplanted donor cells, the

regulatory factors, and the cell signaling pathways leading to the recruitment of host

cells, have been thoroughly reviewed.

3.2 Cell-based therapy for bone defects

The treatments for a multitude of skeletal deficits arising from tumors, infections,

trauma, biochemical disorders, and abnormal skeletal development have been

conducted clinically through the development of surgical techniques for autologous

bone graft, allogeneic tissue transplantation and the implementation of biocompatible

materials [48]. However, each of these reconstructive options has its disadvantages.

The limited availability of autologous bone graft that can be harvested, and

considerable morbidity at the donor site remains inherent limitations of autologous

transplantation [49]. Conversely, although the use of allograft or xenograft avoids

impairments to the donor site, it is sometimes associated with the risks of infection,

and unfavorable immune response from the host [50, 51]. In addition, certain bone-

substitute materials can lead to poor integration, adverse reactions and eventual bone

resorption [52, 53]. Recently, encouraging results from in vitro [54] and in vivo [55,

56] studies, as well as early human clinical applications [57, 58], have aroused intense

interest in cell-based therapy as an alternative to the traditional techniques for bone

defect repair and regeneration.

Page 29: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

9 | P a g e

Figure 1. Schematic of cell-based therapy strategy. The cell transplantation/delivery strategy

involves tissue biopsies, ex vivo expansion of cells from the biopsies, and subsequent transplantation of

the cells into the recipient. Adapted from [59].

The traditional concept of cell-based therapy involves the harvest of suitable cell

sources from autologous/allogenic donors, ex vivo cultivation and expansion to a large

population (Figure 1). These cells are either transplanted back into to the patient by

injection, or seeded onto a three-dimensional scaffold and then implanted into the

recipient [60, 61]. Alternatively, the cell-seeded scaffold can be incubated in a

bioreactor, where the cells are exposed to certain biological, chemical or physical

stimuli that promote the formation of the appropriate tissue prior to the implantation

[62, 63]. The rationale for cell-based therapy to induce bone regeneration is based on

the high osteogenic potency of osteoinductive cells, which may actively induce

adjacent progenitor cells to form new bone tissue [64]. Recent investigations probing

progenitor cell populations have produced a large amount of evidence to support the

clinical viability of cell-based bone tissue engineering [25]. Most of the studies have

focused on the selection of cell sources that are safe, less costly and more easily

available. However, a key unresolved question is whether these cells themselves

participate in the regenerative process mainly by differentiating into osteoblasts or

facilitating tissue repair through the production of various trophic factors (cytokines)

that may modulate the host tissue microenvironment. This may facilitate the

differentiation of resident stem cells and lead to the cell recruitment through blood

vessels, bringing even more of the host cells to the bone defect sites. Recent research

Page 30: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

10 | P a g e

has also revealed that donor cell number [65], nutrition and matrix acidity in the

recipient site [66, 67] can also have different effects on the regenerative process.

3.3 Cell sources for cell-based bone tissue engineering

3.3.1 Mesenchymal stromal cell (MSC)

Regardless of their original sources, stem cells share two characteristic properties.

Firstly, they have the capacity for prolonged or unlimited self-renewal under certain

conditions, and secondly they retain the potential to differentiate into a variety of

more specialized cell types [68]. Undoubtedly, human embryonic stem cells (hESCs)

are able to differentiate down an osteogenic lineage in an efficient manner and

maintain an adequate proliferative capacity post-harvest [69]. However, the political

and ethical issues associated with hESCs have cast a shadow of doubt over their

realistic potential for implementation in tissue engineering strategies [70]. Thus,

MSCs have emerged as a promising cell source in the ongoing research of bone tissue

engineering [71]. Numerous studies have demonstrated that MSCs derived from

various origins, including skin, hair follicle, periosteum, bone marrow, adipose tissue,

and umbilical cord blood, are able to differentiate into osteoprogenitors and

osteoblasts in vitro and in vivo [72-77]. The osteogenic capacity of MSCs can be

further enhanced by the systemic or local administration of bone morphogenic

proteins (BMPs) or other osteoinductive biomolecules, either in slow-release systems

or viral/non-viral based gene delivery systems [78-82]. The addition of supporting

biomolecules has gained increasing popularity especially when bone repair in older,

osteoporotic or diabetic individuals is considered, because the differentiation capacity

of autologous MSCs in these conditions is known to be compromised [83-85].

Although the initial enthusiasm surrounding MSC-based bone tissue engineering is

still growing enormously, research towards the goal of defining a cell surface antigen

profile unique to MSCs has only achieved marginal success. According to various

research, MSCs express positive for a long list of cell surface markers, such as CD29,

CD44, CD73, CD90 (Thy-1), CD105, CD117, CD133, CD166 (activated leukocyte-

Page 31: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

11 | P a g e

cell adhesion molecule, ALCAM), and CD186 [86, 87]. Other markers including Sca-

1 (stem cell antigen-1), SCF R/c-kit (stem cell factor), SH2, SH3, SH4, STRO-1, and

HLA Class I have also been identified [88-90]. Furthermore, it is generally agreed

that the immunodepletion of the hematopoietic cells using anti-CD11b, CD34 and

CD45, forms the fundamental concept for negative selection method of MSCs

purification [91, 92].

In addition, the systemic analysis of cell surface molecules of MSCs has revealed that

MSCs express receptors for numerous extracellular matrix (ECM) proteins including

collagen (α1β1-, α2β1-integrin), laminin (α6β1-, α6β4-integrin), fibronectin (α3β1-,

α5β1-integrin) and vitronectin (αvβ1-, αvβ3-integrin) [93-96]. These specific

expression patterns of adhesion molecules suggest the potential interactions between

MSCs and other cell types in vivo [97-99]. Owing to the complexity of the stem cell

niche, the study of dynamic interactions between stem cells and differentiated

neighbouring cells in their physiological microenvironment has only recently been

explored [100-103]. Depending on the ECM and signal molecules in the

microenvironment, MSCs can develop into adipocytes, osteoblasts, chondrocytes,

myoblasts or other phenotypes [104-107]. However, no single marker has yet been

identified that definitively delineates MSCs in vivo and hence there is a lack of

thorough understanding of the mechanisms underlying stem cell renewal and its

functional differentiation characteristics.

3.3.2 Bone marrow stromal cell (BMSC)

The first MSC population to gain significant attention from researchers and provide

the largest contribution to the current knowledge regarding the osteogenic potential of

MSCs is derived from the bone marrow cavity, which is in tight contact with the

hematopoietic compartment [108]. Bone marrow stromal cells (BMSCs) have been

identified as a population of organized hierarchical postnatal stem cells with the

potential to undergo osteogenic, as well as chondrogenic and adipogenic

differentiation when exposed to appropriate microenvironmental cues [76].

Page 32: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

12 | P a g e

Because of their multipotent nature, BMSCs have become one of the most important

cell sources for cell-based therapy and tissue regeneration [109]. Their osteogenic

differentiation potential has been well characterised in many in vitro studies [110-

112]. In addition, animal model-based studies also reveal that BMSCs have

significant capability for healing bone defects of critical size in vivo when implanted

with proper biomaterials [113, 114], indicating their great potential for bone tissue

engineering applications. In terms of human studies, the use of in vitro expanded

autologous BMSCs in conjunction with porous hydroxyapatite ceramic scaffolds has

been reported in the treatment of large bone defects [115]. Progressive integration of

the implants with the native bone was observed as well as new bone formation inside

the bioceramic pores and vascular ingrowth. A favorable integration of the implants

with the pre-existing bone was maintained during the long-term follow-up period and

no major adverse reactions were found, illustrating the durability achieved by the cell-

based bone tissue engineering approach [116].

Osteogenic induction may enhance the probability of in vivo bone formation and

contribute to the reliability of bone tissue engineering. Several recent investigations

demonstrated that in vitro osteogenic induction of BMSCs before implantation could

significantly promote subsequent ectopic bone formation compared to

undifferentiated BMSCs [19]. Therefore, it may be feasible to obtain a relatively

small amount of autologous BMSCs from patients, expand and pre-differentiate them

into an appropriate number of osteoblastic cells and implant them back to the donors

to facilitate the repair and regeneration of bone defects. Recent research has also

addressed the importance of a proper microenvironment for BMSC transplantation

[117]. It is hypothesized that injured tissues produce the appropriate signals necessary

for cell transplantation and that the local microenvironment may eventually induce

particular phenotypes and functions of the transplanted cells [118-120]. However, the

specific microenvironment (i.e., the unique combination of matrix, growth factors,

and cell adhesion cues), which is critically important for the control of progenitor cell

maintenance, proliferation, and differentiation, and is required to determine the final

fate of the transplanted BMSCs in vivo, remains unknown.

Page 33: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

13 | P a g e

3.3.3 Osteoblast (OB)

Understanding bone growth regulation, the continued remodeling of bone throughout

life, and the regeneration of injured tissues are the basic biological concerns of

clinicians for the treatment of skeletal diseases. The major event that triggers

osteogenesis is the transition of MSCs into bone forming osteoblasts (OBs). OBs are

mononuclear, not terminally differentiated cells that are responsible for the synthesis,

deposition and mineralization of ECM. Therefore, they play a pivotal role in creating

and maintaining skeletal architecture. MSCs need to undergo several transitional steps

before becoming mature OBs. Each transition requires complex activation or

suppression of critical molecular elements for the commitment and differentiation of

OBs to occur (Figure 2).

Figure 2. Schematic of osteoblast differentiation. Adapted from [121].

Over the last decade, molecular and genetic studies have broadened our understanding

of OB differentiation. Ducy and Karsenty conducted the initial study on the regulation

of osteocalcin, which is the most specific gene of OBs [122]. The earliest

osteoprogenitor cells express Runt-related transcription factor 2 (RUNX2), a bone

Page 34: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

14 | P a g e

specific transcription factor, which is required for the early stages of OB

differentiation [123]. Osteoblastogenesis consists of three major stages, proliferation,

matrix maturation, and mineralization, which are characterised by sequentially

expressed distinctive markers from OBs. Alkaline phosphatase (ALP), type I collagen

(COLI), osteocalcin (OCN), osteopontin (OPN) and PTH-related protein (PTHrP)

receptor are the most frequently used markers during osteoblastogenesis [124, 125].

Generally, ALP and COLI are early markers of OBs’ differentiation, while OCN and

PTHrP receptors appear late. OPN reaches its peak during proliferation and in the

later stages of differentiation.

Following the initial lineage commitment, there is a phase of expansion. The initial

cell division is asymmetric, giving rise to another stem cell and a committed

osteoprogenitor. The pre-osteoblasts are at an intermediate stage. They still proliferate

and then develop into OBs that synthesize bone matrix. The mature OBs lie adjacent

to the newly formed osteoid and express ALP, OPN, BSP, and OCN. The cumulative

effect of the recruitment of stem cells and their expansion, and the functional capacity

of mature OBs, are measured by rates of bone formation in vivo. There are three fates

upon completion of the synthetic phase in the terminal stage of the remodeling cycle.

OBs may become osteocytes upon entrapment within the mineralized matrix, evolve

into inactive lining cells that protect the bone matrix from osteoclasts, or undergo

apoptosis [126].

Immature OBs derived from MSCs may provide more consistency in forming bone

than precursor populations with multipotentiality in terms of cell-based therapy for

bone defect healing [127, 128]. However, the isolation procedure of OBs is

considerably labour-intensive and time consuming. In addition, only a relatively small

number of cells are available after the dissociation of the tissue and their expansion

rates are comparatively low. Consequently, the generation of clinically adequate

numbers of OBs for cell-based therapy is a significant challenge.

Page 35: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

15 | P a g e

3.4 The contribution of donor cells in cell-based bone tissue

engineering

Although the outcome of cell-based therapy using the aforementioned cell sources has

been encouraging, the precise mechanisms by which the donor cells enhance tissue

repair and regeneration are not clearly understood. In most cases, the intent of cell-

based therapy is to repair damaged tissues with donor cells capable of carrying out

structural and functional recovery [128]. However, the extent of donor cell

contribution to the cells/tissues of interest has not been fully elucidated. It becomes

apparent that in many situations, the donor cells improve tissue regeneration at the site

of damage without significant engraftment or differentiation [129]. Studies in which

MSCs were transplanted into children with severe forms of osteogenesis imperfecta

(OI) revealed the level of donor cell engraftment was extremely low and could not

justify the growth acceleration and the bone fracture rate reduction [130-132]. In other

related studies, transplantation of BMSCs into animal models of myocardial infarction

(MI) led to significantly improved cardiac function due to the secreted factors from

donor cells, rather than direct engraftment or differentiation [133, 134]. These

findings lead to the speculation that besides direct cell-to-cell contact, the donor cells

may secrete proinflammatory cytokines (such as IL-1 and IL-6), mitogens (such as

IGF, FGF and PDGF), morphogens (BMPs), and angiogenic factors (VEGF and

angiopoietins) [135, 136], which exert paracrine activities and modulate the

microenvironment for neighboring stem cells to carry out the reparative process [137-

140]. In addition, the broad spectrum of bioactive molecules that are produced by the

donor cells may lead to the tropism of endogenous cells from their original niches.

In the field of cell-based bone tissue engineering, a recent study from Cancedda’s

team has shed some light on the commitment of donor cells by using a murine model

of ectopic bone formation. This particular study demonstrated that the origin of newly

formed bone was dependent on the maturation status of the implanted cells [141]. An

intramembranous ossification directly performed by the donor cells was observed

when mature OBs were seeded onto a porous ceramic. In contrast, the implantation of

Page 36: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

16 | P a g e

undifferentiated MSCs led to the formation of new bone of host origin via

endochondral ossification. Researchers have also shown that MSCs are able to

accelerate the wound healing process by stimulating cell differentiation [142], and

releasing pro-angiogenic factors that promote new blood vessel formation [143]. The

newly formed vasculature is not only critical for the transportation of oxygen and

nutrients to the centre of the implants, but also fundamental to the recruitment of host

cells through the connection with the host circulating system. This creates a

communicative network between the recipient organism and the engineered graft. In

addition, the implanted cell/scaffold constructs may create a transient inflammatory

microenvironment, in which an innate response is mediated by a mixed cell

population, mainly composed of the recruited monocytes, macrophages, and

lymphocytes. The released factors from the inflammatory cells in turn act as signals

and impact on the way donor cells contribute to the tissue repair and regeneration

process in vivo [144-146]. In this respect, although the molecular mechanisms are yet

poorly understood, it is reasonable to hypothesize that donor cells can act indirectly as

“signaling centres”, orchestrating the host response to the skeletal defects, in addition

to their direct role as osteoprogenitors and osteoblasts.

3.5 The involvement of host cells in cell-based bone tissue

engineering

There is no doubt that cell-based therapy is becoming one of the most promising

strategies to repair irreversibly-affected tissues. However, the therapeutic efficacy

may not only depend on the involvement of donor cells, but also the activation and

homing of endogenous cells from their original niches through the circulation to the

injured tissues. Within a complex niche, endogenous cells are exposed to a variety of

spatially and temporally controlled biochemical mixtures of insoluble transmembrane

receptor ligands, proteases, adhesion molecules (such as selectins and integrins),

ECM molecules, as well as soluble chemokines, cytokines and growth factors [147,

148]. In the field of stem cell biology, the term “homing” is often defined as the

recruitment of endogenous cells to the damaged tissues or their navigation to other

targeted anatomic destinations following mobilization [149], often over great

Page 37: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

17 | P a g e

distances via the bloodstream. In addition, a highly proliferative population of

progenitor cells residing in the neighboring healthy tissues may also be recruited to

the defect sites [59] (Figure 3).

Figure 3. Schematic of endogenous cells homing to the damaged tissues for in situ

tissue regeneration. The homing of endogenous cells involves actively recruiting host stem cells

from either the blood or a tissue specific niche to the site of injury. Adapted from [59].

Host cell mobilization is not only essential in a wide range of physiological and

pathological phenomena, but also important to biotechnological applications such as

regenerative medicine and tissue engineering. Previous work has demonstrated that

endogenous biological resources and environmental conditions can be used for in situ

tissue regeneration, suggesting the possibility to reduce the intensive labour for donor

Exit tissue

niche to

enter blood

Page 38: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

18 | P a g e

cell procurement and manipulation [150]. Applications of in situ tissue regeneration

are primarily based on the local release of bioactive molecules, such as cytokines and

growth factors, to enhance the mobilization of host cells and control the fate of the

recruited endogenous cells for efficient tissue regeneration. Mao and colleagues have

successfully fabricated a TGFβ3-infused scaffold which could induce homing of

endogenous cells to regenerate the entire articular surface of the rabbit synovial joint

[151]. A more recent study instead emphasized the efficiency of combined systemic

and local delivery of stem cell recruiting factors for tissue engineering, showing that

the combined system could significantly enhance the recruitment of host cells which

were capable of differentiating into multiple lineages [152].

Likewise, in cell-based bone tissue engineering, a continuous source of cell

recruitment to the bone defect site is ensured by the endogenous cells. The research

by Tasso and coworkers showed that two consecutive waves of host cell migration

were induced by the donor cells seeded on the porous ceramic scaffolds [153]. The

first wave, observed 7 days after the implantation, was enriched in CD31+ endothelial

cells, whereas the second wave, observed at day 11, was enriched in CD146+

perivascular cells, potentially responsible for the newly formed bone. Another study

has also demonstrated that osteoblastic precursors can be systemically recruited from

remote bone marrow cavities via the bloodstream and become involved in fracture

healing [154].

The full comprehension of the host response to implanted donor cells is of great

importance to the development of novel clinical strategies where host cells could

contribute to regenerate the appropriate tissue. What remains unclear is the

mechanism of how the endogenous cells are recruited to the damaged

tissue/transplantation site, and what potential role the recruited host cells play in the

process of tissue repair and regeneration. Therefore, it is necessary to further

understand the phenomenon of host/donor cells interaction, which may benefit the

optimization of treatment alternatives for nonunion fractures and other pathologic

skeletal deficits.

Page 39: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

19 | P a g e

3.6 Chemokines and growth factors related with cell

migration

Cell migration is a fundamental component of embryogenesis, vasculogenesis, wound

healing, immune surveillance, as well as tumor cell metastasis. It is a mechanically

integrated molecular process regulated by a series of coordinated mechanisms [155].

Cells become asymmetrical and polarized during migration, with front and back ends

formed to direct movement [156]. Broad lamellipodia and spike-like filopodia take

place primarily around the cell front, whereas, stress fibers, which are involved in cell

adhesion, are formed or deformed at the back [157]. These multi-step processes

involve morphological changes that result from the rearrangement of actin

cytoskeleton, the emergence of protrusive membrane structures followed by the

contraction of the cell body, the detachment of the uropod and the secretion of matrix

degrading enzymes [157, 158]. A multitude of chemokines, cytokines and growth

factors play critical roles in regulating the mobilization and homing of MSCs for

effective regeneration of functional tissue.

3.6.1 CXCL12

Chemokines (Chemotactic cytokines) are a family of small (8-10 kDa) proteins able to

chemically direct responsive cells, such as lymphocytes, neutrophils, hematopoietic

cells and other cell types to the targeted tissue compartment [159]. In addition,

chemokines are thought to play an important role in cell activation, differentiation and

survival [160-162]. Members of the chemokines are divided into four subfamilies,

namely CXC, CC, C and CX3C chemokines, depending on the spacing of their first

two cysteine residues [159, 163]. Of these, CXC and CC chemokines are the two

major groups.

Chemokine (C-X-C motif) ligand 12 (CXCL12) is the most predominant stem cell

homing factor to date [164]. It was first identified as stromal cell-derived factor-1

Page 40: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

20 | P a g e

(SDF-1) in the supernatant of BMSCs [165]. CXCL12 is strongly expressed in bone

marrow and produced by osteoblasts, endothelial cells and reticular cells that are

scattered through the bone marrow stroma [166]. CXCL12 is highly conservative and

has 99% homology between mouse and human, allowing CXCL12 to act across

species barriers. Upon stimulation with CXCL12, MSCs are able to rearrange the

actin cytoskeleton and increase phosphorylation of the focal adhesion adapter protein

paxillin, which is required for focal adhesion turnover and ultimately migration [167].

The critical role for CXCL12 in the homing of endogenous cells to damaged sites has

been unraveled by studies in animal models of liver, limb, and heart damage [168-

171]. One of the most important functions of CXCL12 is related to tissue repair and

regeneration. Recent investigations have shown the repair of ischemic injuries

involves the selective recruitment of circulating or resident progenitor cells. Hypoxia-

inducible factor-1 (HIF-1), a central regulator of tissue hypoxia, induces CXCL12

expression in ischemic areas in direct proportion to reduced oxygen tension in vivo

[172, 173]. The elevated CXCL12 then recruits the circulating progenitor cells to

repair the damaged tissues.

Chemokine (C-X-C motif) receptor 4 (CXCR4) is the best characterised receptor for

CXCL12 and an essential mediator for the chemotaxis responses induced by CXCL12

in many responsive cells [174]. Previous studies have shown the CXCR4-deficient

mice died in utero or perinatally due to multiple defects in developing hematopoietic

and nervous system [175-177], leading to the conclusion that CXCR4 is indispensible

to several physiological processes during late embryonic and early postnatal stages. A

recent study has also provided evidence that CXCR4 functions in postnatal bone

development by regulating OB development in cooperation with BMPs signaling,

indicating CXCR4 acts as an endogenous signaling component necessary for bone

formation [178]. In bone marrow, CXCL12 availability has been shown to be

regulated by the uptake of CXCL12 via CXCR4 positive cells, which then translocate

the chemokine into the bone marrow. This transporter function is characteristic of

both endothelial and stromal cells [179]. The binding of CXCL12 to CXCR4 leads to

receptor structural changes and G-protein activation, which then stimulates signaling

Page 41: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

21 | P a g e

cascades (e.g., PI3K and MAPKs), and induces intracellular Ca2+

release, resulting in

cell migration, proliferation and survival (Figure 4).

The interaction of CXCL12 with CXCR4 is thought to regulate trafficking of the

hematopoietic stem cells, endothelial progenitor cells (EPCs) and in prostate and

breast cancer metastasis to bone and lung [180, 181]. The pivotal role of the

CXCL12/CXCR4 axis in the mobilization and homing of stem cells to injured or

stressed tissue is further investigated by numerous researchers [182-184]. Previous

work has demonstrated that applying a neutralizing CXCR4 antibody after induction

of cerebral ischemia in a rat model compromised BMSC homing, resulting in poor

neurological recovery after stroke [185]. On the other hand, transfection or

transduction of MSCs with CXCR4 is reported to enhance cell migration toward

CXCL12 in vitro [186, 187] and toward infracted myocardium in rats [188], whereas

almost no migration is observed to normal myocardium [188]. Recent studies have

emphasized the clinical effect of CXCR4 antagonist. The bicyclam AMD3100, also

known as Plerixafor (Mozobil™), is a competitive selective CXCR4 antagonist. It

blocks the binding of CXCR4 to CXCL12 and has been demonstrated to interfere

with a number of pathophysiological processes [189], such as rheumatoid, allergic

and malignant diseases. Physiologically, bone marrow has a higher CXCL12

concentration than any other tissues, therefore, circulating MSCs home to bone

marrow proficiently. AMD3100 has been shown to mobilize CD34+ stem cells from

the bone marrow into the blood circulation and augment the migration of bone

marrow-derived EPCs into sites of neovascularization after myocardial infarction

[190]. Currently, AMD3100 is actively pursued as a stem cell mobilizer for

autologous transplantation in patients with multiple myeloma and non-Hodgkin's

lymphoma [189], and has great potential for the treatment of other hematological

malignancies and non-hematological malignancies.

Page 42: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

22 | P a g e

Figure 4. CXCL12/CXCR4 signaling pathway. Binding of CXCL12 to CXCR4 leads to

receptor structural changes and G-protein activation. Activated G-proteins induce multiple signaling

pathways, such as PI3K, MAPKs and NFκB, and the release of intracellular Ca2+. These signaling

pathways are associated with cell proliferation, migration and survival.

3.6.2 VEGF

As most members from the VEGF/platelet-derived growth factor (PDGF) family,

VEGF is the potent mitogen for endothelial cells (ECs), and one of the key regulators

of angiogenesis, vasculogenesis and developmental hematopoiesis. In addition to its

well-documented effects on blood vessel development and formation [191, 192],

VEGF also plays a critical role in promoting osteogenesis [193]. VEGF is of great

importance for the patterning of the bone growth plate [194, 195] by stimulating the

Page 43: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

23 | P a g e

differentiation of OBs [196] and the crosstalk between OBs and ECs [197]. In a

mouse femur fracture study, treatment with exogenous VEGF enhanced not only

blood vessel formation but also ossification and new bone formation, whereas VEGF

inhibition impaired callus mineralization [198]. Various cell types secrete VEGF,

including tumor cells, macrophages, OBs and osteoblast-like cells, and several growth

factors, hormones, and cytokines regulate its expression. It has been demonstrated that

VEGF secretion from osteoblastic cells is at low levels in the early stage of osteogenic

differentiation [199] and markedly increases as osteoblastogenesis proceeds [200].

This is significantly regulated by OB-specific transcription factor Osterix, as reported

in a recent study [201]. Interestingly, VEGF has been proved to be chemoattractive

for primary human osteoblasts, providing first evidence that it may also be involved in

local recruitment of osteoprogenitor cells in the course of endochondral bone

formation or remolding [202, 203].

There are five structurally related mammalian VEGF ligands, namely, VEGF-A,

VEGF-B, VEGF-C, VEGF-D and placental growth factor (PlGF) [204]. Each VEGF

ligand has several different variants due to either alternative splicing or processing.

Being the most abundant and active member of the VEGF family, VEGF-A

undergoes alternative splicing, which results in at least six isoforms (VEGF-A121,

VEGF-A145, VEGF-A165, VEGF-A183, VEGF-A189 and VEGF-A206), among which

VEGF-A165 is the most predominant and biologically potent. The VEGFs are

produced by many different cell types and typically act locally in a paracrine manner.

However, VEGF-A produced by ECs may act in an autocrine manner to stimulate

vessel survival [205].

The VEGFs bind to three structurally related VEGFR tyrosine kinases, VEGFR-1

(Flt-1), VEGFR-2 (KDR/Flk-1) and VEGFR-3 (Flt-4), each of which forms a

homodimer on ligand binding [206] (Figure 5). VEGF-A binds to VEGFR-1 and

VEGFR-2, but not VEGFR-3. The majority of VEGF signaling is mediated through

VEGFR-2 in endothelial cells [206, 207]. VEGF-A165 can also interact with

neuropilin-1 (NRP-1) and neuropilin-2 (NRP-2), which are cell surface

Page 44: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

24 | P a g e

transmembrane glycoprotein receptors [208]. NRPs have a short cytoplasmic domain,

thus it is speculated that they do not mediate the signal independently, but function

only as co-receptors [208]. It has been reported that NRP-1 can associate and form a

complex with VEGFR-2, leading to an enhanced VEGFR-2 signaling induced by

VEGF-A165 binding to NRP-1 [209]. One interesting finding is that VEGF-A can

stimulate MSCs proliferation and mobilization to damaged tissues by binding to and

activating PDGF receptors, even though MSCs express low basal level of VEGFRs

[210].

Figure 5. Vascular endothelial growth factor (VEGF) family members and their

receptors. The mammalian VEGF family consists of five structurally related glycoproteins, VEGF-A,

VEGF-B, VEGF-C, VEGF-D and placental growth factor (PlGF). The VEGFs bind to three

structurally related VEGFR tyrosine kinases. VEGF-A binds to both VEGFR-1 and VEGFR-2. VEGF-

B and PlGF bind exclusively to VEGFR-1. VEGFR-3 is a specific receptor for VEGF-C and VEGF-D.

VEGF-C and VEGF-D can be proteolytically processed to allow binding to VEGFR-2 as well. In

addition, neuropilin (NRP)-1 and NRP-2 act as co-receptors for specific isoforms of VEGF family

members, and increase the binding affinity of these ligands to their respective receptors. Adapted from

[211].

Page 45: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

25 | P a g e

3.6.3 PDGF

PDGFs are the primary mitogens for the cells of mesenchymal and neuroectodermal

origin and are members of the evolutionarily conserved, structurally and functionally

related VEGF/PDGF family [212]. First identified in 1970s as a serum growth factor

that stimulates proliferation of fibroblasts, smooth muscle cells (SMCs) and glial cells

[213-215], the PDGF family is now one of the best characterised growth factor

systems.

The PDGF family consists of four different isoforms, the traditional PDGF-A and

PDGF-B, and more recently discovered PDGF-C and PDGF-D [216]. PDGF-A and

PDGF-B are processed intracellularly and are secreted in their active forms, while

PDGF-C and PDGF-D are secreted as latent factors and require extracellular cleavage

of the CUB domain. This is carried out by the tissue-type plasminogen activator (tPA)

for PDGF-C, and the urokinase-type plasminogen activator (uPA) or matriptase for

PDGF-D [217-220]. The PDGF ligands exist as disulphide-bonded polypeptide chains,

including the four homodimers PDGF-AA, PDGF-BB, PDGF-CC and PDGF-DD,

and one heterodimer, PDGF-AB [221]. The ligands exert their biological activities via

two distinct but structurally related, membrane bound receptor tyrosine kinases

(RTKs), PDGF receptor-α (PDGFR-α) and PDGF receptor-β (PDGFR-β). Particularly

strong expression of PDGFR-α has been noticed in subtypes of mesenchymal

progenitors in lung, skin and intestine, as well as in oligodendrocyte progenitors,

while PDGFR-β is expressed in the mesenchyme, especially in vascular SMCs

(vSMCs) and pericytes [222]. Ball et al. also reported that MSCs in vitro have a high

cell surface ratio of PDGFR-α/PDGFR-β [223], and PDGFR-α appears to be a

characteristic of undifferentiated MSCs. The PDGFRs have common domain

structures, including five extracellular immunoglobulin (Ig) loops and a split

intracellular tyrosine kinase domain [222]. The binding of ligand to the receptor

triggers receptor homodimerization or heterodimerization, resulting in

autophosphorylation of specific tyrosine residues within the intracellular domains and

kinase activation. Once activated, intracellular mediators dock to phosphotyrosine

residues in the receptor, leading to the activation of downstream signaling pathways,

e.g. PI3K, MAPK and PLC-γ. The ability of each of the five dimeric PDGF ligands to

Page 46: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

26 | P a g e

bind and activate the PDGF receptors is quite specific as summarized in Figure 6.

PDGF-AA can only interact with PDGFR-α, whereas PDGF-BB has a greater binding

affinity for PDGFR-β. However, PDGFR-β is also capable of binding PDGFR-α, and

thus can induce all three receptor homodimer and heterodimer. PDGF-AB and PDGF-

CC can both bind to PDGFR-α, and initiate the formation of homodimer and

heterodimer. PDGF-DD binds predominantly to PDGFR-β and forms a homodimer

PDGFR-ββ, but also may induce lower levels of the heterodimeric PDGFR-αβ [222,

224].

Figure 6. Simplified schematic of the platelet-derived growth factor (PDGF) system.

PDGF-A and-B are secreted as active homo- or heterodimers. PDGF-C and –D are secreted as

homodimers and latent factors which need extracellular cleavage of the CUB domain by tPA (PDGF-

C), uPA (PDGF-D) or matriptase (PDGF-D) for receptor binding and activation. PDGF-AA is the most

selective member of the PDGF family and activates PDGFR-αα exclusively. PDGF-BB is the universal

ligand. PDGF-AB and PDGF-CC assemble and activate PDGFR-αα and PDGFR-αβ. PDGF-DD

activates PDGFR-ββ, and under certain circumstances, PDGFR-αβ. PDGF binding results in

autophosphorylation and activation of different signaling pathways, such as JAK/STAT, PI3K, PLC-γ

or MAPK. Downstream signaling promotes gene expression and mediates the biological functions of

the PDGF isoforms, e.g. proliferation, migration, and survival. Adapted from [225].

Page 47: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

27 | P a g e

Since being originally isolated due to its ability to promote cell proliferation, PDGF

has been appreciated to initiate additional cellular responses such as cell survival and

migration. PDGF signaling is undoubtedly required for the spreading of various

populations of cells during embryonic development, such as oligodendrocyte

precursors in the spinal cord, neural crest mesenchymal cells toward the branchial

pouches and cardiac outflow tract, and pericytes along newly formed angiogenic

sprouts [222]. The mechanisms by which PDGF regulates the process of cell

spreading remain largely undefined in most of the situations. However, recent studies

in Drosophila, Xenopus, and mouse have demonstrated a role for PDGF family

members in directed cell migration in certain developmental processes in vivo. PDGF-

AA induced chemotaxis is well documented [226], and PDGFR-α promotes a

chemotactic response in various cell types [227]. Nagel et al. demonstrated PDGF-

A/PDGFR-α signaling is required for directional migration of mesoderm on its

endogenous matrix substratum, as well as for cell orientation in the embryo [228].

Knockout of the genes encoding PDGFR-β or PDGF-B in mice causes deficient mural

cell recruitment, leading to widespread microvascular bleeding [229-231]. PDGF-BB,

the main ligand of PDGFR-β, is a potent stimulant of SMCs recruitment during

neointimal hyperplasia following vascular injury [222]. PDGFR signaling has also

emerged as a predominant pathway in the recruitment of perivascular MSCs, which

play fundamental roles in angiogenesis, wound repair and tissue regeneration [210,

232-234]. A recent study has further identified the synergistic relationship between

α5β1-integrin and PDGFR-β. Cell adhesion to fibronectin induced PDGFR-β

phosphorylation in an α5β1-integrin dependent manner, leading to the activation of

the PI3K/Akt pathway, actin reorganization and recruitment of MSCs [235].

3.7 Signaling pathways related with cell migration

In order to participate in tissue repair and regeneration, MSCs have to be mobilized

and migrate to the target sites and integrate with the local tissues. Understanding the

potential signaling pathways in relation to the endogenous cell homing can offer new

insights into the management of bone defect healing.

Page 48: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

28 | P a g e

3.7.1 PI3K/Akt signaling pathway

Phosphoinositide 3-kinases (PI3Ks) were initially discovered as lipid kinases

associated with viral oncoproteins about 20 years ago [236]. Since then, the

association between PI3Ks and cancer has been further elucidated [237]. Over the last

decade, growing evidence has indicated that preferential activation of PI3Ks is

important for defining the leading edge in a wide range of motile cell types [238-240].

PI3K has been the focus of attention with respect to its activation by chemokine

receptors and the role it plays in regulating cell migration [241, 242]. The PI3K

family can be divided into three main classes according to their in vitro lipid substrate

specificity, enzyme structure and products formed [243]. The heterodimeric class I

PI3Ks consist of two structurally closely related subgroups. Class IA enzymes share

one of the common regulatory subunits (p85α and -β, p50α, and p55α and -γ) and

different catalytic subunits (p110α, -β, and -γ) [244]. Class IB enzymes comprises a

p101 or p84 regulatory subunit as well as the p110γ catalytic isoform [245, 246].

Classes IA and IB are the most extensively studied PI3Ks and, together with their

lipid product phosphatidylinositol (3,4,5)-triphosphate (Ptdlns(3,4,5)P3), have been

widely implicated in controlling cell migration and polarity [247] (Figure 7). Class II

PI3Ks lack a regulatory subunit and contain three isoforms (C2α, -β, and -γ), which

are capable to phosphorylate Ptdlns and Ptdlns(4)P to Ptdlns(3)P and Ptdlns(3,4)P2,

respectively. Recent studies have suggested that class II PI3Ks may play a positive

role in the regulation of cell adhesion and actin reorganization during cell migration

and wound healing in non-immune cell systems [248, 249]. The sole member of class

III PI3K is vacuolar protein sorting 34 (Vps34), which consists of a regulatory subunit,

p150, for its activity. Vps34 produces only Ptdlns(3)P from Ptdlns and is involved in

multiple steps of membrane trafficking. However, no defined role of Vps 34 in cell

migration has been reported so far [250].

Page 49: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

29 | P a g e

Figure 7. Signaling through phosphatidylinositol (3,4,5)-triphosphate. On stimulation of

class I PI3Ks together with their lipid product phosphatidylinositol-3,4,5-trisphosphate

(Ptdlns(3,4,5)P3), several phosphatases are involved in subsequent dephosphorylation steps (dashed

arrows). PTEN hydrolyses the 3-phosphate of Ptdlns(3,4,5)P3 to regenerate phosphatidylinositol-4,5-

bisphosphate (Ptdlns(4,5)P2), thus terminating PI3K signaling. 5-phosphatases such as SHIP, however,

generate Ptdlns(3,4)P2 that, like Ptdlns(3,4,5)P3, can facilitate the activation of Akt. The key reaction

steps outlined are integrated into complex signaling networks in which Akt and other targets of 3-

phosphorylated phosphoinositides, together with further downstream components, regulate cell growth,

proliferation, survival and motility. Adapted from [251].

The serine/threonine kinase Akt, also known as protein kinase B (PKB), is the best

characterised downstream effector of class І PI3Ks. The Akt family consists of three

isoforms, namely Akt1 (PKBα), Akt2 (PKBβ) and Akt3 (PKBγ). They are expressed

ubiquitously, share similar activation mechanisms and typically initiate signal relay to

secondary signaling cascades by phosphorylating an overlapping subset of particular

proteins [252]. Akt has emerged as one of the key regulators for cell survival,

proliferation, angiogenesis and glucose metabolism [253-256]. A growing body of

evidence indicates that Akt may also affect cytoskeletal changes and promote cell

migration in different organisms [257-260]. However, the role of isoform-specific Akt

family members in the regulation of cell migration has been difficult to ascertain.

Activation of Akt1 has been found to negatively influence mammary epithelial cell

migration and epithelial-mesenchymal transition (EMT) [261], whereas siRNA

knockdown of Akt1, but not Akt2, results in a considerable increase in cell migration,

Page 50: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

30 | P a g e

which was related to the activation of extracellular-signal-regulated kinase-1 and -2

(ERK1/2) [261]. The inhibitory effect of Akt1 on cell motility and invasion has also

been investigated in various breast cancer cell lines, in which degradation of the

nuclear factor of activated T cells (NFAT) transcription factors was involved [262].

On the contrary, other reports have claimed a distinctive role of Akt1 in promoting

cell migration and invasion [263, 264]. Similarly, the positive effect of Akt2 on cell

motility has been well established. Recent investigations have demonstrated that the

reduction of Akt2 inhibits chemotaxis signal transduction in glioma cells [265] and

breast cancer cells [266]. On the other hand, Akt2 has been found to be a negative

regulator of Pak1 [267]. Akt2 knock-out mouse embryonic fibroblasts have increased

migration, especially invasion through ECM, suggesting a negative role of Akt2 in

regulation of Rac/Pak signaling, the actin cytoskeleton and cell migration [267].

Interestingly, an inhibitory role for both Akt1 and Akt2 in prostate cancer migration

and invasion has been reported in a recent study [268]. However, the inconclusive

effects of Akt1 and Akt2 on cancer cell migration may not translate to other cell types.

These studies demonstrate the importance of crosstalk between the PI3K/Akt pathway

and other pathways and highlight the cell type-specific actions of Akt kinases in the

regulation of cell motility.

Several PI3K/Akt stimulators have been documented to enhance cell migration [269-

272]. The role of CXCL12, one such stimulator, in cancer cell invasion has been well

studied. Binding of CXCL12 to CXCR4 leads to the stimulation of a signaling

cascade that involves activation of multiple targets, including ERK1/2, p38 and Akt

[273-276]. It has been reported previously that CXCL12 could stimulate a rapid and

transient accumulation of PtdIns(3,4,5)P3 in leukaemic T cell lines and peripheral

blood-derived lymphocytes within 15 sec, and the elevated accumulation returned to

basal level 5 min after chemokine treatment [277]. Other work has further

demonstrated the cell migration induced by CXCL12 can be abrogated by the PI3K

inhibitors wortmannin and LY294002 [241, 242, 273]. Hypoxia pre-conditioning has

also been shown to enhance MSCs migration and improve their tissue regenerative

potential via the activation of the PI3K/Akt signaling pathway [278].

Page 51: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

31 | P a g e

A number of phosphorylation targets for Akt are now emerging. Enomoto and

colleagues have recently identified a novel substrate of Akt, designated Girdin

(girders of actin filaments), which is an actin binding protein [279]. The accumulation

of phosphorylated Girdin at the leading edge of migrating cells suggests that Girdin is

essential for the remodeling of the actin cytoskeleton that propels the cells forward

and provides a direct link between Akt and cell motility [279, 280]. The p70 S6

kinase (p70S6K

) is also a downstream effector of the PI3K/Akt pathway, which

regulates cytoskeleton dynamics via the actin filament cross-linking proteins. The

overexpression of p70S6K

in ovarian cancer cells has been reported to induce a marked

reorganization of the actin cytoskeleton and promote directional cell migration [281].

Researchers have demonstrated that p70S6K

can stimulate the rapid activation of Rho

family small GTPases Cdc42 and Rac1, as well as their major downstream effector,

p21-activated kinase 1 (PAK1) [281, 282]. PAK1 is the first discovered member of

the mammalian PAK family, which comprises six proteins divided in group І (PAK1-

3) and group II (PAK4-6) according to their structural and functional features [283,

284]. PAK1 plays a fundamental role in controlling cell motility by linking a variety

of extracellular signals to changes in actin cytoskeleton organization, cell shape and

adhesion dynamics [285-287].

3.7.2 MAPK signaling pathway

Mitogen-activated protein kinases (MAPKs) belongs to a large family of highly

conserved serine/threonine protein kinases, which are found in all eukaryotic cells,

and expressed in virtually all tissue types. To date, six distinct classes of MAPKs have

been characterised, namely, the extracellular signal-regulated kinases (ERK) 1 and 2

(ERK1/2), ERK3/4, ERK5, ERK7/8, p38 (α/β/γ/δ) MAPK, and c-Jun N-terminal

kinases (JNK1/2/3) [288-290]. JNK1/2/3 and p38 (α/β/γ/δ) MAPK are collectively

called stress-activated protein kinases (SAPK). ERK5 is also known as big MAPK

(BMK1) [291, 292]. Very little is known about the atypical MAPK enzymes, ERK3/4

and ERK7/8. However, all the members of MAPKs consist of a homodimer of

molecular mass of about 80 kDa and contain the signature sequence T–X-Y, where T

Page 52: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

32 | P a g e

and Y are threonine and tyrosine, and X denotes glutamate in ERK, proline in JNK,

and glycine in p38, respectively [293, 294].

MAPKs are pivotal components of a complex intracellular signal transduction

pathway that functions to integrate the cells’ response to extracellular stimuli (Figure

8). The activation of MAPKs proceeds through a series of upstream molecules in an

orderly fashion. Various extracellular stimulations, e.g. hormones, growth factors,

cytokines and stress signals, are relayed from the cell surface through an elegant

cascade of phosphorylation events leading to the cell nucleus, where a diverse array of

intracellular responses occur [295]. In general, ERK1/2 are preferentially activated in

response to growth factors that act through receptor tyrosine kinases, e.g. epidermal

growth factor (EGF), fibroblast growth factor (FGF) and PDGF, whereas the

JNK1/2/3 and p38 (α/β/γ/δ) MAPKs are more responsive to pro-inflammatory

cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β or stress

stimuli ranging from osmotic shock to ionizing radiation [289, 290, 295].

Consistent with their critical roles in key cellular activities, including cell proliferation,

differentiation, survival and apoptosis [296], the MAPK signaling pathway has also

been implicated in the migration of various cell types [297]. Accumulating data

suggest that MAPKs are essential in regulating the dynamics of focal adhesion and

the reorganization of microtubules and filamentous actin that is required for cell

morphogenesis and migration [297]. ERK1/2 is the most extensively studied

subfamily of MAPKs and has been implicated in the migration of a wide range of cell

types. Numerous studies have reported the ERK pathway inhibitors PD98059 and

U0126 suppress cell migration in response to ECM, such as fibronectin, vitronectin

and collagen [298, 299], growth factors, such as EGF, FGF, PDGF and VEGF [300-

303], and other stimuli, such as fetal bovine serum and urokinase-type plasminogen

activator (uPA) [304, 305].

Page 53: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

33 | P a g e

Figure 8. Schematic of the three mammalian mitogen-activated protein kinase

(MAPK) signaling pathways. Extracellular stimuli activate the MAPK pathways through

mechanisms mediated by GTPases, including RAS, RAC, CDC42 (cell-division cycle 42) and RHO

(RAS homologue). Once MAPKKKs (MAPK kinase kinases), such as RAF, MEKK (MAPK/ERK

kinase kinase) and TAK (TGFβ-activated kinase), are activated, they phosphorylate MAPKKs (MAPK

kinases) on two serine residues. MAPKKs in turn phosphorylate the MAPKs ERK (extracellular-

signal-regulated kinase), JNK (JUN N-terminal kinase) and p38 on both threonine and tyrosine

residues, which results in the catalytic activation of these MAPKs. Activated MAPKs can translocate to

the nucleus to phosphorylate a number of transcription factors, such as ternary complex factor (TCF)

family members and components in the activator protein 1 (AP1) complexes, including JUN and

activating transcription factor 2 (ATF2), thereby altering gene transcription. TCF forms a complex with

serum response factor (SRF) to regulate Fos induction. AP1 is involved in the transcription of a wide

variety of genes. Adapted from [306].

Page 54: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

34 | P a g e

3.8 Summary

Although small bone defects may heal through the body’s normal reparative process,

large skeletal deficits overwhelm this system and may fail to reach a satisfied

structural and functional recovery, despite the use of conventional reconstructive

approaches. With the recent advanced research in regenerative medicine and tissue

engineering, cell-based therapy has emerged as one of the most promising approaches

for tissue repair and regeneration. Cell-cell interaction between host and donor cells is

believed to play a critical role in enhancing the healing process after in vivo cell

transplantation. However, little is known about how donor cells lead to tissue repair

and regeneration after transplantation or how they interact with the host cells in vivo.

In addition, a comprehensive understanding of the host response to the transplanted

donor cells is crucial for developing novel clinical strategies based on the active host

cell contribution to the regeneration of damaged tissues. The recruitment process of

endogenous cells may be facilitated by external stimuli, such as the manipulation of

donor cells and the application of homing factors, which may offer new therapeutic

options to harness the host’s innate capacity for the repair and regeneration of bone

defects. Migration of MSCs is controlled by complicated signal networks.

Understanding the molecular mechanisms of MSC migration will help to develop

strategies to facilitate the tissue forming capacity of cell delivery therapy and to

optimize cell-based bone tissue engineering.

Page 55: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

35 | P a g e

CHAPTER 4 RESEARCH REPORTS

Page 56: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

36 | P a g e

Page 57: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

37 | P a g e

REPORT ONE

Implantation of osteogenically differentiated donor mesenchymal

stromal cells causes recruitment of host cells

Yinghong Zhou, Wei Fan, Indira Prasadam, Ross Crawford, Yin Xiao

Suggested Statement of Contribution of Co-Authors for Thesis by

Published Papers:

Contributors Statement of contribution*

Yinghong Zhou Involved in the conception and design of the project, performed

laboratory experiments, data analysis and interpretation. Wrote

the manuscript.

Wei Fan Involved in sample collection, and assisted with manuscript

preparation.

Indira Prasadam Assisted with manuscript preparation.

Ross Crawford Involved in the conception and design of the project, assisted in

collection of clinical samples, and reviewed the manuscript.

Yin Xiao Involved in the conception and design of the project, and

reviewed the manuscript.

Principal Supervisor Confirmation

I have sighted email or other correspondence from all co-authors confirming

their certifying authorship.

Name Signature Date

Page 58: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

38 | P a g e

ABSTRACT

Background: The interaction between host and donor cells is believed to play an

important role in bone tissue engineering. However, it is still unclear how donor cells

behave and interact with host cells in vivo. The purpose of this study was to track the

interactions between transplanted osteogenic cells and donor cells during osteogenesis.

Methods: An in vitro migration assay was carried out to investigate the ability of

osteogenically differentiated human mesenchymal stromal cells (O-hMSCs) to recruit

undifferentiated mesenchymal stromal cell (MSCs). At the in vivo level, O-hMSCs

were implanted subcutaneously or into skull defects in severe combined

immunodeficient (SCID) mice. New bone formation was observed by µCT and

histological procedures. In situ hybridisation (ISH) against human Alu sequences was

performed to distinguish donor cells from host cells.

Results: The in vitro migration assay revealed an increased migration potential of

MSCs by co-culturing with O-hMSCs. In agreement with the results of in vitro studies,

ISH against human Alu sequences showed that host mouse MSCs migrated in large

numbers into the transplantation site in response to O-hMSCs, suggesting that donor

cell characteristics may help to recruit the host MSCs for transplantation success.

Interestingly, host cells recruited by O-hMSCs were the major cell populations in

newly formed bone tissues, indicating that O-hMSCs can trigger and initiate

osteogenesis when transplanted in orthotopic sites.

Conclusions: The observations from this study demonstrated that in vitro induced O-

hMSCs were able to attract host MSCs in vivo and were involved in the osteogenesis

together with host cells, which may be of importance for bone tissue engineering

applications.

Page 59: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

39 | P a g e

INTRODUCTION

Rapid expansion of knowledge regarding progenitor cell-based tissue engineering

offers new insights into the treatment of various kinds of diseases [24], especially

those related to bone defects as a result of tumour, infection, trauma, biochemical

disorders, and abnormal skeletal development [307]. Cell transplantation is one of the

most promising strategies used to enhance the tissue-healing process, as it introduces

functioning cells directly into affected areas [308, 309]. The concept of cell-based

therapy involves cell isolation from autologous, allogenic, or xenogenic sources, cell

proliferation and differentiation in vitro, and application in vivo [23, 310, 311].

Recently, this concept has been tested in large bone defect healing in several studies

[39, 312].

Cell-cell interactions between host and donor cells are believed to play an important

role in enhancing the healing process after in vivo cell transplantation [313]. In cell

therapy for bone defect healing, the delivery of osteogenic cells into the defect area is

often considered to be of direct help to the new bone formation [314]. Due to the

reduced multipotency compared with progenitor cells, osteoblasts may be more

committed to form new bone tissue [64]. A study on the rabbit mandible showed that

transplantation of osteoblast-like cells has the potential to promote maturity of the

distracted callus [315]. Another study reported that transplantation of bone marrow

stromal cells contributed to new bone formation in vivo [316]. However, until now,

cell therapy for bone tissue engineering has focused on determining a source of

exogenous cells with osteogenic capacity, without paying attention to the possibility

that the recipient itself might provide the cell elements necessary for tissue repair.

Little evidence shows how and to what extent the transplanted donor osteogenic cells

behave and interact with host mesenchymal stromal cells (MSCs) during the new

bone formation process [317]. The purpose of this study was to investigate how

transplanted osteogenically differentiated human MSCs (O-hMSCs) were involved in

osteogenic processes and how they interact with host MSCs after transplantation.

Page 60: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

40 | P a g e

MATERIALS AND METHODS

Cell culture

Human bone marrow was sourced from patients (n=5, three females and two males)

in the Department of Orthopaedics at Prince Charles Hospital, with informed consent

and ethics approval from the Ethics Committee of Queensland University of

Technology. The average age of the patients involved in this study was 69.6 ± 3.7

(range 65-75) years. The hMSCs for this study were isolated by density gradient

centrifugation over Lymphoprep™ (Axis-Shield PoC AS, Oslo, Norway) according to

the manufacturer’s protocol [318, 319]. The cells were then seeded in culture flasks in

Dulbecco’s modified Eagle’s medium (DMEM; Gibco®, Life Technologies Pty Ltd.,

Australia) containing 10% (v/v) fetal bovine serum (FBS; In Vitro Technologies,

Australia) and 1% (v/v) penicillin/streptomycin (P/S; Gibco®, Life Technologies Pty

Ltd., Australia). Following 3 days incubation in a humidified atmosphere of 5% CO2

at 37ºC, the non-adherent cells were washed away leaving behind the adherent cell

population that was growing in clusters. Upon reaching 70-80% confluence, the

attached hMSCs were subcultured after treatment with 0.25% trypsin (Gibco®, Life

Technologies Pty Ltd., Australia) and 1mM EDTA (Gibco®, Life Technologies Pty

Ltd., Australia). In vitro osteogenic differentiation of hMSCs was performed in

osteogenic medium (DMEM supplemented with 10% FBS, 1% P/S, 10 mM β-

glycerophosphate, 50 µM ascorbic acid and 100 nM dexamethasone; Sigma Aldrich,

Australia) for 2 weeks.

Characterisation of hMSCs

To confirm the phenotype of hMSCs obtained through in vitro culture, flow

cytometric analysis against the cell surface markers CD34, CD45, CD73, CD90 and

CD105 (BD Biosciences, North Ryde, NSW, Australia; Clontech, Palo Alto, CA,

USA) was performed [320]. Briefly, after incubation with primary antibodies against

the aforementioned cell markers and repeated washing with 3% PBS/bovine serum

albumin (BSA), hMSCs were resuspended and incubated with secondary antibody

(phycoerythrin-conjugated goat anti-mouse immunoglobulin G; Jackson

Page 61: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

41 | P a g e

ImmunoResearch Laboratories, West Grove, PA, USA), then resuspended in 3%

PBS/BSA and read through in a flow cytometry machine (Becton-Dickinson, Cowley,

Oxford, UK). In the negative control, the primary antibody was omitted and replaced

by a mouse isotype IgG.

In vitro migration assay

hMSCs (1×104) were seeded on polyethylene terephthalate (PET) track-etched

membrane transwell inserts (10 mm effective diameter of membrand with 8 µm pore

size; Becton-Dickinson Labware, Franklin Lakes, NJ, USA). After 24 h of cell

attachment, the transwell inserts were assembled onto the 12-well companion plates

cultured with 1×104 hMSCs, which were subjected to osteogenic differentiation (O-

hMSCs) or retained in undifferentiated condition (hMSCs) for 2 weeks prior to the

migration assay. All co-cultures were performed in serum-free medium to rule out the

effect of serum in complete medium on the migration of hMSCs. Cell migration was

allowed to proceed for 8 h. Cells that had migrated to the other side of the insert

membrane were fixed, and then stained with crystal violet. Images were taken with an

inverted microscope (Eclipse TS100, Nikon Australia Pty Ltd.). The migration was

quantified by counting the number of cells that migrated through the pores in six

randomly selected fields on each transwell insert membrane, and values from three

independent experiments, with triplicate samples in each experiment, were averaged

and showed as mean ± SD.

In vitro osteogenic differentiation of hMSCs on collagen scaffolds

Collagen scaffolds were fabricated from a 1mm thick bovine type I collagen sheet

(Medtronic Australia Pty Ltd, North Ryde, NSW, Australia), using a hole puncher 5

mm across and sterilized with 75% ethanol, followed by three rinses of PBS prior to

cell seeding. For the in vitro osteogenic differentiation, 6×104

hMSCs were first

seeded onto the collagen membrane and cultured in osteogenic medium for 2 weeks.

Collagen with no cells and collagen with undifferentiated hMSCs were used as

controls.

Page 62: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

42 | P a g e

In vivo implantation of osteogenically differentiated hMSCs

Nine 5 week-old male severe combined immunodeficient (SCID) mice (Animal

Resources Centre, Canning Vale, WA, Australia) were used for the skull defect model

[78, 321], and another three SCID mice were used for subcutaneous cell implantation.

For the skull defect implantation, an incision was made longitudinally along the

centre line of the shaved skull skin. A 2.5mm diameter defect was then made through

the skull, using a dental burr. The defect was treated as follows with three mice in

each group: i.e. defects filled with collagen scaffold, collagen carrying

undifferentiated hMSCs, or collagen carrying O-hMSCs. For the subcutaneous cell

implantation, three longitudinal incisions were made in each animal along the central

line of the shaved dorsum, approximately 1 cm apart. Each individual pocket received

one scaffold from each group, using a complete randomized design for cell-free

collagen scaffolds, collagen scaffolds carrying hMSCs, or collagen scaffolds carrying

O-hMSCs. The SCID mice were sacrificed 2 weeks after subcutaneous implantation

and 4 weeks after the skull defect implantation.

Micro-computed tomography (μCT) assessment

All the samples harvested were fixed in 4% paraformaldehyde overnight at room

temperature and then washed in PBS. Cross-sectional scans parallel to the cell-

seeding surface of the implants were performed in a µCT scanner (µCT40, Scanco

Medical AG, Brüttisellen, Switzerland) at a resolution of 12 µm and three-

dimensional (3D) images of the implants were reconstructed. The calcified area inside

the implant on each scanned slice was circled out and the volume of calcification

within the implants was measured using the inbuilt scanner software package and

recorded for statistical analysis.

Histology and immunohistochemistry (IHC)

All harvested subcutaneous samples were embedded in paraffin blocks for sectioning,

while the skull defect samples were first decalcified in 10% EDTA solution for 4

Page 63: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

43 | P a g e

weeks and then embedded in paraffin. 5 µm serial sections were cut from the

embedded samples, using a microtome (Leica Microsystems GmbH, Wetzlar,

Germany). Only sections close to the centre of the samples were used for the staining.

For haematoxylin and eosin (H&E) staining, tissue slices were dewaxed in xylene and

rehydrated in descending concentrations of ethanol (100%, 90% and 70%), then

stained with Mayer’s haematoxylin (HD Scientific Supplies Pty Ltd., NSW, Australia)

for 2 min before rinsing with tap water for 5 min. The slides were dehydrated in

ascending concentration of ethanol (70%, 90% and 100%) then stained with eosin

(HD Scientific Supplies Pty Ltd., NSW, Australia) for 15 s, cleared with xylene and

mounted with DPX mounting medium (Fronine, Laboratory Supplies, NSW,

Australia).

IHC staining was performed for the assessment of new bone formation in the skull

defects. In brief, sections were blocked with 10% swine serum for 1 h, then incubated

with primary alkaline phosphatase antibody (ALP; 1:400, anti-human; Sigma-Aldrich,

Australia) overnight at 4°C. The sections were washed free of primary antibody and

incubated with biotinylated secondary antibody (Dako, CA, USA) for 15 min,

followed by incubation with horseradish peroxidise (HRP)-conjugated streptavidin-

biotin complex (Dako, CA, USA) for 15 min. Antibody complexes were visualized by

the addition of diaminobenzidine (DAB) for 3 min. The sections were then lightly

counterstained with haematoxylin and mounted with mounting medium.

von Kossa staining

In order to observe the calcification in subcutaneous implants, samples were subjected

to 5% silver nitrate (Sigma-Aldrich, Australia) and exposed to light for 30 min. The

sections were then washed in distilled water and dehydrated in ethanol before being

mounted with cover slips and mounting medium.

Page 64: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

44 | P a g e

In situ hybridisation (ISH)

To trace the donor cells (i.e O-hMSCs) in skull defect sites and subcutaneous implants,

human Alu probe was obtained from PanPath (Budel, the Netherlands). Briefly,

hybridisation of the Alu probe was carried out for 16 h at 37°C in a humidified

chamber after denaturation at 95°C for 5 min, followed by post-hybridisation washing

procedure. 3-Amino-9-ethylcarbazole (AEC) was then applied to visualize the

hybridisation. The sections were then counterstained with haematoxylin, and mounted

with Crystal MountTM

aqueous mounting medium (Sigma-Aldrich, Australia).

The localisation of transplanted human cells identified by the Alu probe in both

subcutaneous and skull defect scaffolds was observed, recorded and analysed. The

lining cells around the initial bone were counted as osteoblasts and those trapped in

the bone matrix and isolated in lacunae were counted as osteocytes. Three sections

from each group were included and the cell percentage of both donor human cells and

host mouse cells in three randomly selected areas on each section within the implant,

newly-formed bone tissues and bone marrows adjacent to the implants or from the

femur was calculated and recorded for further analysis.

Statistical analysis

Non-parametric Kruskal-Wallis test was performed to distinguish the differences

among all the groups in the in vitro migration assay and the µCT quantified data

analysis. The differences of cell percentage between donor and host cells inside

samples were subjected to Wilcoxon test. Significant difference was considered at

p<0.05.

Page 65: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

45 | P a g e

RESULTS

hMSCs characterisation and in vitro osteogenic differentiation

To verify the cells derived from the human bone marrow are MSCs, a panel of

antibodies against stem cell markers was chosen to evaluate the phenotypes of the

hMSCs used in the present study. Flow cytometry against CD34, CD45, CD73, CD90,

and CD105 revealed that the in vitro expanded hMSCs were negative for

haematopoietic cell surface markers of CD34 and CD45, while positive for selected

mesenchymal cell surface markers of CD73, CD90 and CD105 (Figure 9A). The

hMSCs derived from the bone marrow showed typical fibroblastic morphology

(Figure 9B). von Kossa staining showed calcium precipitation by hMSCs after in

vitro osteogenic differentiation, confirming the mineralization potential of hMSCs

derived from bone marrow (Figure 9C).

Figure 9. hMSCs characterisation and in vitro osteogenic differentiation. (A) Flow

cytometric analysis of cell surface antigen expression in hMSCs showed negative expression of

haematopoietic cell surface markers (CD34 and CD45) and positive expression of mesenchymal cell

surface markers (CD73, CD90 and CD105). (B) Microscopic observation of undifferentiated hMSCs

showed typical fibroblastic morphology. (C) von Kossa staining showed the mineralization potential of

hMSCs after osteogenic differentiation.

Page 66: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

46 | P a g e

In vitro migration assay

The effect of O-hMSC on the migration of hMSCs was assessed using a transwell

device containing a cell culture insert with 8 µm pores. After 8 h of incubation, the

number of hMSCs which migrated through the transwell membrane increased

significantly in the co-culture with O-hMSCs (Figure 10A, D, G) compared with the

serum-free medium (Figure 2A, B, E) and the co-culture with undifferentiated hMSCs

(Figure 10A, C, F) (p<0.05).

Figure 10. In vitro migration assay. (A) Comparison of the number of hMSCs that migrated to

the other side of the insert membrane among different groups. SF, serum-free medium; hMSC,

undifferentiated hMSCs were seeded in the companion plate; O-hMSC, osteogenically differentiated

hMSCs were seeded in the companion plate (*p<0.05). (B)-(D) Schematics representation of co-culture

groups. (E)-(G) Images of hMSCs that migrated to the other side of the insert membrane: (E) Serum-

free medium; (F) hMSCs were seeded in the companion plate; and (G) O-hMSCs were seeded in the

companion plate.

Page 67: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

47 | P a g e

Osteogenesis in subcutaneous and skull defect implants

H&E staining on ectopic implants showed no histological signs of new bone

formation (Figure 11I-K). However, µCT and von Kossa staining showed

mineralization along collagen fibres in the scaffolds carrying O-hMSCs (Figure 11C,

D, L). There was no mineralization in the scaffolds carrying undifferentiated hMSCs

or in the cell-free scaffolds (Figure 11A, B, D).

H&E staining revealed significant new bone formation, showing a trabecular bone-

like structure within the implants carrying O-hMSCs in the skull defect areas (Figure

11O). µCT scanning and 3D reconstruction revealed that a substantial amount of

mineralization was found in the implants carrying O-hMSCs, showing the greatest

volume of the calcified areas (Figure 11G, H). However, there was almost no new

bone formation within the cell-free or undifferentiated hMSCs-carrying implants in

the skull defect areas (Figure 11E, F, H, M, N). The newly formed bone in the O-

hMSCs implants was further confirmed by IHC staining against ALP (Figure 11P).

Cell migration in ectopic sites

In the subcutaneous implants, ISH against human Alu sequences revealed that host

mouse cells (Alu-negative) migrated into the centre of the implants, gathered together

and were then surrounded by donor O-hMSCs. This showed a close relationship

between the transplanted human cells and the host mouse cells in the osteogenic

process (Figure 12D). On the other hand, in the cell-free or undifferentiated hMSCs-

loaded scaffolds (Figure 12B, C), the host mouse cells distributed sparsely in the

transplanted scaffolds. A higher host:donor cell ratio was found inside the scaffolds

when O-hMSCs were implanted subcutaneously (Figure 12A; p<0.05).

Page 68: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

48 | P a g e

Figure 11. Osteogenesis in subcutaneous and skull defect implants. (A)-(C) 3D images of

subcutaneous implants with scaffold only, scaffold+hMSCs or scaffold+O-hMSCs. (D) Quantification

of the bone volume of the calcified areas within the subcutaneous implants (*p<0.05). (E)-(G) 3D

images of skull defect treated with scaffold only, scaffold+hMSCs or scaffold+O-hMSCs. (H)

Quantification of bone volume of the calcified areas within the skull defect implants (*p<0.05). (I)-(K)

H&E staining of the subcutaneous implants showed no new bone formation in all three groups. (L) von

Kossa staining revealed calcification within the subcutaneous implants carrying O-hMSCs. H&E

staining of skull defect filled with scaffold alone (M) and scaffold+hMSCs (N), showing no new bone

formation. (O) H&E staining of skull defect filled with scaffold+O-hMSCs showing new bone

formation. (P) ALP staining revealed active osteoblasts in the new bone tissues in skull defect treated

with scaffold +O-hMSCs.

Page 69: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

49 | P a g e

Figure 12. Distribution of donor human cells and host mouse cells in the

subcutaneous implants revealed by ISH. (A) Quantification and comparison of host:donor cell

ratio between the hMSCs and O-hMSCs groups hMSC, undifferentiated hMSCs; O-hMSC,

osteogenically differentiated hMSCs (*p<0.05). (B) Implants with collagen scaffold only. (C) Implants

with hMSCs showing both the donor (red, arrow) and host cells (blue, arrowhead). (D) Implants with

O-hMSCs showing both the donor (red, arrow) and host cells (blue, arrow head).

Cell migration in orthotopic sites

In the skull defect areas, ISH against human Alu sequences revealed that more than

60% of total cells in the observed areas were host mouse cells within the O-hMSCs

implants (Figure 13A; p<0.01). In newly formed bone tissues, O-hMSCs were found

to turn into both osteoblasts and osteocytes (Figure 13D-F). However, the number of

human-originated osteoblasts and osteocytes were significantly less than the cells

from the host, indicating more host cells were involved in the newly formed bone

tissues (Figure 13B-F; p<0.05).

Page 70: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

50 | P a g e

Alu-positive cells in the newly formed bone marrow

In the bone marrow of the newly formed bone within the implants carrying O-hMSCs,

Alu-positive cells were identified coexisting with host marrow cells and occupied

about 30% of total bone marrow cells (Figure 13H). Some Alu-positive cells were

round in shape and mixed with Alu-negative host cells in the newly formed bone

marrow, while others were found attached to the endosteal bone surfaces (Figure

13G). In the bone marrow close to the defect, no Alu-positive cells were found

(Figure 13I).

To investigate whether transplanted donor cells migrated into the circulation system,

femur samples from the same animal after cell transplantation were collected and

processed. No Alu-positive cells were found in bone marrow collected from the femur

samples, indicating that transplanted O-hMSCs were only involved in local

osteogenesis (Figure 13J).

Page 71: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

51 | P a g e

Figure 13. Distribution of O-hMSCs and host mouse cells in the skull defect implants

and bone marrows, revealed by ISH. (A)-(C) Quantification and comparison of cell percentage

between donor and host cells (O-hMSC: osteogenically differentiated hMSCs): (A) General cell

percentage; (B) Cell percentage as osteoblasts; (C) Cell percentage as osteocytes (*p<0.05, **p<0.01).

(D)-(F) Skull defect implants showed both the donor (red) and host cells (blue); arrows, donor cells-

derived osteoblasts on new bone surface; arrowhead, donor cells-derived osteocytes. (G) Bone marrow

in skull defect showing both donor human cells (red) and host mouse cells (blue); arrow, marrow cell-

like transplanted donor cells; arrowhead, endosteal cell-like transplanted donor cells. (H)

Quantification and comparison of cell percentage between donor O-hMSCs and host mouse cells in

bone marrow (*p<0.05). (I) Bone marrow adjacent to the skull defect. (J) Bone marrow in the femur of

the mouse with cell-transplanted skull defect.

Page 72: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

52 | P a g e

DISCUSSION

Cell therapies have tremendous potential to treat a wide array of diseases, and the

major strategy of cell transplantation is via scaffolds, which are intended to serve as

templates for tissue formation [322]. However, integration of templated tissues with

host tissues remains a significant challenge. Since MSCs were first isolated from bone

marrow [323], they have been considered to be among the most important stem cell

sources for cell therapy, due to their multilineage differentiation capacity [116, 324,

325]. Although the osteogenic differentiation potential of MSCs has been well

characterised in many in vitro and in vivo studies [110, 326-328], no study has been

conducted so far to clarify how the donor cells interact with the host cells in the

osteogenic process.

In the present study, the in vivo behaviour of O-hMSCs was investigated using the

ISH technique against human Alu sequences in both subcutaneous (ectopic) and skull

defect (orthotopic) osteogenesis. The results showed that O-hMSCs played a

significant role in host cell recruitment in both ectopic and orthotopic sites. The cell

distribution and the ratio of donor:host cells were significantly different in ectopic and

orthotopic sites.

Subcutaneous transplantation of O-hMSCs revealed that the host cells migrated into

the centre of O-hMSCs and exhibited almost a 50:50 donor:host cell ratio. This

phenomenon may indicate that ectopic osteogenesis of transplanted O-hMSCs needs

the involvement of a certain amount of host cells to generate an optimal local

microenvironment for initial calcification. The host cells formed cell clusters or

nodules among the transplanted cells, indicating close cell-cell communication

between host and donor cells.

In the skull defect areas, a substantial of newly formed bone tissue was found in

implants carrying O-hMSCs. It is very interesting to find that these transplanted O-

Page 73: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

53 | P a g e

hMSCs were involved in the new bone formation and differentiated to both

osteoblasts and osteocytes at a similar donor:host cell ratio. Our finding of a large

number of recipient cells at the defect sites confirmed cell migration into the defect.

More host cells than donor cells differentiated into osteoblasts and osteocytes in skull

defect areas. This is different from the subcutaneous implant, which showed a nearly

equivalent number between the host and donor cells. This difference might be

attributed to the different local environment (ectopic and orthotopic) and different

osteogenic process (calcification and new bone formation). The smaller number of O-

hMSCs compared with host cells as osteoblasts and osteocytes in skull defect areas

may indicate that transplanted donor cells played a more important role in initiating

osteogenesis, rather than in maintaining the bone formation process. Different

proliferation rates between the human and mouse cells, postoperative complications

present at the defect sites, and other factors, such as immunological reactions, may

also tip the balance in favour of the recipient cells.

Another interesting aspect of tracking the transplanted donor cells is to investigate the

role of donor cells in the reconstitution of bone marrow in the newly formed bone,

and whether they could enter the blood flow and circulate into the bone marrow cell

compartment. In this study, only in the bone marrow cavities in the skull defects were

the donor cells found. This means that at the 4 week time point, the donor cells could

recruit host haematopoietic cells to form bone marrow in newly formed bone.

However, donor cells did not travel to the far end of body extremities. In the new

bone marrow, transplanted cells took up a lower cell percentage than the host marrow

cells. Considering the similarly low percentage of donor O-hMSCs as osteoblasts and

osteocytes in newly formed bone tissues, in vivo osteogenesis seems to require more

involvement of host cells than donor cells. In the bone marrow, some transplanted

cells presented the round shape of host marrow cells, but some were attached to the

endosteal bone cavity surface. This phenomenon suggests some transplanted cells

could become endosteal cells.

Page 74: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

54 | P a g e

The reason why O-hMSCs recruited more MSCs compared to undifferentiated

hMSCs is not clear. It is possible that cytokines released by O-hMSCs may have

stimulated MSCs to travel toward O-hMSCs. A recent study demonstrated that SDF-

1, a member in the chemokine family, is important for the migration of MSCs to bone

marrow [164]. Many studies have shown that during tissue repair, local SDF-1

expression is upregulated and acts as a signal to recruit the CXCR4 (SDF-1 ligand)

expressing cells from circulation and/or bone marrow. Apart from chemokines, other

molecular factors, such as fibroblast growth factor (bFGF) [329], matrix

metalloproteinases (MMPs) [330], and TLR ligation [331] were known to increase the

migratory homing activity of MSCs to the damaged tissue. It is possible that one or

more of the factors described above, or other factors unknown, are induced by O-

hMSCs, giving rise to a positive feedback of the host MSCs, which in turn trigger the

bone formation.

This study demonstrated that in both ectopic and orthotopic sites, transplanted O-

hMSCs could recruit host cells to initiate osteogenesis involving the formation of

osteoblasts, osteocytes, and bone marrow components. The ratio of donor:host cells

inside the transplants were significantly dependent on the local environment and

directly correlated with osteogenesis. This new finding provides the groundwork for

future studies about the interaction between transplanted donor cells and host cells in

bone defect healing.

ACKNOWLEDGEMENTS

The authors declare that there is no conflict of interest and gratefully acknowledge the

contribution of Ms Wei Shi to this study. The work was supported by Queensland

University of Technology.

Page 75: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

55 | P a g e

REPORT TWO

VEGF induced CXCL12/CXCR4 axis in the recruitment of mesenchymal

stromal cells (MSCs) during osteogenesis

Yinghong Zhou, Wei Fan, Indira Prasadam, Ross Crawford, Yin Xiao

Suggested Statement of Contribution of Co-Authors for Thesis by

Published Papers:

Contributors Statement of contribution*

Yinghong Zhou Involved in the conception and design of the project, performed

laboratory experiments, data analysis and interpretation. Wrote

the manuscript.

Wei Fan Involved in sample collection, and assisted with manuscript

preparation.

Indira Prasadam Assisted with manuscript preparation.

Ross Crawford Involved in the conception and design of the project, and assisted

in collection of clinical samples.

Yin Xiao Involved in the conception and design of the project, and

reviewed the manuscript.

Principal Supervisor Confirmation

I have sighted email or other correspondence from all co-authors confirming

their certifying authorship.

Name Signature Date

Page 76: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

56 | P a g e

ABSTRACT

Background: Cell-cell interaction is believed to play important roles in the cell-based

therapy for bone defect healing. However, it is still unclear what factors are involved

in the interaction between donor cells and host cells during the bone healing process.

The purpose of this study was to investigate the potential effect of vascular

endothelial growth factor (VEGF) on the expression pattern of CXCL12/CXCR4 with

respect to the interactions between undifferentiated and osteogenically differentiated

mesenchymal stromal cells (MSCs and OMSCs) in vitro and in vivo.

Methods: Indirect co-culture models were applied to study the effect of VEGF

derived from OMSCs on the motility, the cell morphology and the CXCL12/CXCR4

expression of MSCs in vitro. The role of VEGF in the regulation of the host cell

recruitment was monitored by the application of neutralizing VEGF antibody in vivo,

after OMSC implantation into the skull defects in SCID mice.

Results: Our data showed that VEGF secretion increased dramatically in MSCs after

osteogenic differentiation in vitro and the secretion of VEGF promoted the motility of

MSCs with elongated actin filament and enhanced the expression of

CXCL12/CXCR4. In vivo findings revealed remarkable new bone formation within

the defects implanted with OMSCs. Furthermore, blocking VEGF with neutralizing

antibody resulted in significant decrease of MSCs recruitment and new bone

formation.

Conclusions: The observations from this study demonstrate that VEGF secreted by in

vitro induced OMSCs plays a pivotal role in MSC recruitment possibly via the

activation of the CXCL12/CXCR4 axis during osteogenesis.

Page 77: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

57 | P a g e

INTRODUCTION

Bone defect remains one of the most prevalent clinical problems even though various

strategies to stimulate bone healing have been explored [78, 332-335]. Due to

morbidity issues in donor sites and the limited availability of autografts, allograft

transplantation has become an attractive alternative treatment option. However, the

clinical results are limited and sometimes associated with poor clinical predictability

and severe adverse reactions [336]. In order to overcome these limitations, it is of

great interest to develop novel biological strategies, which require elucidation of the

molecular signals responsible for successful bone regeneration.

Cell-based therapy, such as mesenchymal stromal cell (MSC) transplantation, is a

promising strategy for the treatment of bone defects. MSCs are attractive candidates

for cell-based bone regeneration [337], because of their capacity to differentiate into

osteogenic lineage following in vitro expansion and osteogenic induction. The

development, remodelling and repair of bone are controlled by the proliferation,

differentiation, migration and apoptosis of various cell types, including osteoblasts,

osteoclasts and haematopoietic cells [338]. Although a lot of studies have focused on

the ability of implanted cells to differentiate within the defect sites, more recent

research suggests other important factors may be involved in the bone tissue

regeneration, such as the regulation of direct or indirect cell-cell interactions [339,

340], as well as the release of soluble factors [341]. However, the paracrine factors

and molecular mechanisms that contribute to the interaction between host cells and

donor cells during osteogenesis are poorly understood.

Recent studies have shown that cellular movement and relocalisation are essential for

many fundamental physiologic properties, including angiogenesis, chemotaxis, and

osteogenesis [342, 343]. Osteogenesis and angiogenesis are two closely correlated

processes during bone formation and regeneration [344]. Vascular endothelial growth

factor (VEGF) is highly expressed in vascularised tissues and can promote endothelial

cell proliferation, differentiation and migration from pre-existing vasculatures. As the

best characterised angiogenic mediator, VEGF is believed to play a crucial role in

skeletal development by enhancement of angiogenesis [345, 346]. On the other hand,

Page 78: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

58 | P a g e

a key feature regulating cell movement is chemotaxis, which induces migration via

signaling molecules termed chemokines [347, 348]. Chemokines are a large family of

small, basic polypeptides that have mainly been characterised as chemoattractants.

They navigate trafficking and localisation for various cell type in tissue compartments

[349]. The interaction between chemokine (C-X-C motif) ligand 12 (CXCL12) and its

receptor CXCR4 is believed to be the most important contributor to MSC homing to

areas of tissue damage and is involved in tissue regeneration [350-354]. However,

there is little information to be found regarding the potential effect of VEGF on the

expression pattern of CXCL12/CXCR4 with respect to the recruitment of MSCs

during new bone formation. We therefore investigated whether VEGF secretion by

osteogenically differentiated MSCs (OMSCs) was associated with CXCL12/CXCR4

expression alterations, which may lead to the migration and recruitment of MSCs in

the osteogenic process.

MATERIALS AND METHODS

Cell culture

Human bone marrow samples were obtained from patients (n=3) undergoing elective

knee replacement surgery in the Department of Orthopaedics at Prince Charles

Hospital with informed consent from each patient and ethics approval from the Ethics

Committee of Queensland University of Technology. Bone marrow was sourced from

the femoral canal. After draining of synovial fluid, a femoral drill hole was made

through the distal femur and an intramedullary rod was passed into the femoral canal

allowing marrow aspirates to be collected with a syringe. Bone marrow-derived

MSCs for this study were isolated by density gradient centrifugation over

Lymphoprep (Axis-shield PoC AS, Oslo, Norway) according to the manufacturer’s

protocol. The cells were then seeded in culture flasks in low glucose Dulbecco’s

Modified Eagle Medium (DMEM; Gibco®, Life Technologies Pty Ltd., Australia)

containing 10% (v/v) fetal bovine serum (FBS; In Vitro Technologies, Australia) and

1% (v/v) penicillin/streptomycin (P/S; Gibco®, Life Technologies Pty Ltd., Australia)

at 37°C, 5% CO2. The medium was changed twice weekly to wash out all non-

adherent cells. Adherent bone marrow-derived MSCs were then passaged once grown

Page 79: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

59 | P a g e

to 70%-80% confluence. Cell characterisation for MSCs has been carried out in our

previous study [318, 355].

Osteogenic differentiation of bone marrow-derived MSCs

Isolated MSCs were subjected to a 2-week osteogenic differentiation in osteogenic

medium (DMEM supplemented with 10% FBS, 1% penicillin/streptomycin, 10 mM

β-glycerophosphate, 50 µM ascorbic acid and 100 nM dexamethasone; Sigma Aldrich,

Australia) for further experiments. Medium was changed twice weekly.

Alizarin Red S staining

In order to identify mineralization nodules, Alizarin Red S staining was performed on

day 14 after MSCs and OMSCs had grown to a confluent monolayer in a 6-well plate.

The medium was removed and the cells were washed with PBS and fixed with 100%

methanol for 10 min at room temperature. After rinsing with distilled water, the cells

were stained in a solution of 1% Alizarin Red S (Sigma-Aldrich, Australia) at pH 4.1

for 20 min and the excessive solution was then washed away. The samples were air

dried and photos were taken under a microscope (Eclipse TS100, Nikon Australia Pty

Ltd.).

Preparation of conditioned medium (CM)

CM was obtained from 1×107

MSCs (CMMSC

) or OMSCs (CMOMSC

). The cells were

washed twice with PBS, and then incubated with 10 mL of serum-free DMEM at

37oC for 48 h. The CM was then collected, subjected to centrifugation at speed of

1000 g for 5 min at 4oC and filtration through a 0.2 µm low protein binding filter

(Millipore Corporation, Billerica, MA, USA) to remove cell debris. The supernatants

were stored at -80oC for subsequent experiments.

Page 80: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

60 | P a g e

Enzyme-linked immunosorbent assay

The concentration of VEGF in CMMSC

and CMOMSC

was tested using ELISA assay kit

for VEGF (R&D Systems Inc., Australia) according to the manufacture’s instruction.

Briefly, all the samples and standards were incubated in the assigned wells of the 96-

well plate for 2 h at room temperature. After three washings, 200 µL VEGF conjugate

was added into each well and incubated for 2 h at room temperature. After washing,

the amount of conjugate bound to each well was determined by the addition of 200 µL

substrate solution. The reaction was quenched by adding 50 µL stop solution per well

and measured immediately at 450 nm with a microplate reader (SpectraMax, Plus 384,

Molecular Devices, Inc., USA). The test was performed in triplicate, and the VEGF

levels were determined using the generated standard curve. All the results were

expressed as the amount (pg) of VEGF in per µL CM.

Indirect co-culture

The CM from MSCs or OMSCs was mixed with fresh culture medium in a ratio of

1:1 before applying to MSCs with or without the addition of 20 ng/mL VEGF

neutralizing antibody (R&D Systems Inc., Australia). The medium was changed every

three days. MSCs were harvested for RNA and protein on day 6 for subsequent

experiments.

In vitro migration assay

MSCs (5×104) were seeded on transwell inserts with 8 µm pore size (Becton-

Dickinson Labware, Franklin Lakes, NJ, USA). After 24 h of cell attachment, the

transwell inserts were assembled onto the 6-well companion plates cultured with

5×104 MSCs, which were subjected to osteogenic differentiation (OMSCs) or retained

in undifferentiated condition (MSCs) for 2 weeks prior to the migration assay. All co-

cultures were performed in serum-free medium with or without the addition of 20

ng/mL VEGF neutralizing antibody. Cell migration was allowed to proceed for 8 h.

Cells that had migrated to the other side of the insert membrane were fixed, and then

Page 81: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

61 | P a g e

stained with crystal violet. Images were taken with an inverted microscope (Eclipse

TS100, Nikon Australia Pty Ltd.). The migration was quantified by counting the

number of cells that migrated through the pores in six randomly selected fields on

each transwell insert membrane, and values from three independent experiments were

averaged and shown as mean ± SD.

xCELLigence system monitored cell migration

The real-time migration tendency of MSCs in response to CMMSC

and CMOMSC

was

further monitored using the CIM-Plate 16 and xCELLigence system RTCA DP

Instrument (Roche Products Pty Ltd., Australia). The CIM-Plate 16 is a 16-well

modified Boyden chamber composed of an upper chamber (UC) and a lower chamber

(LC). The UC is sealed at the bottom by a microporous polyethylene terepthalate

(PET) membrane. These micropores allow the physical translocation of cells from the

upper part of the UC to the bottom side of the membrane, which is covered with

interdigitated gold microelectrode sensors. As migrated cell attach and spread on the

electrode sensors, it leads to an increase in electrical impedance [356]. The LC

contains 16 wells, each of which serves as a reservoir for a chemoattractant solution.

In the present study, 100 µL of cell suspension (4×104 cells) were added to each well

of the UC after serum starvation for 4 h. The UC was then placed on the LC of the

CIM-Plate 16 containing 160 µL CMMSC

and CMOMSC

respectively, with or without

VEGF neutralizing antibody as an attractant. Cell migration was monitored over a

period of up to 12 h. The generated impedance signal was displayed as a

dimensionless parameter termed cell index, which was directly proportional to the

total area of the cell-culture well.

VEGF stimulation of MSCs

In order to further investigate the effect of VEGF on the expression pattern of

CXCL12/CXCR4, cell culture medium supplemented with 20, 40, 80 ng VEGF

(R&D Systems Inc., Australia) per mL was applied to the MSCs. The medium was

replenished every three days through a 6-day culture period, after which the cells were

Page 82: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

62 | P a g e

harvested and subjected to real time quantitative PCR (RT-qPCR) and Western

blotting analysis.

Fluorescent staining for morphological changes

2×103 MSCs were seeded on each coverslip (ProSciTech, QLD, Australia) placed in a

24-well plate. After overnight culture in serum-free medium, CMMSC

, and CMOMSC

with or without VEGF neutralizing antibody, the MSCs were fixed with 4%

paraformaldehyde (PFA) for 30 min. Fixed cells were permeabilized with 0.1% Triton

X-100 in PBS for 5 min and washed twice with 0.1% BSA/PBS. The nuclei were

stained with 0.1 µg/mL DAPI (Life Technologies Pty Ltd., Australia) for 30 min, and

the actin filaments were labelled with rhodamine phalloidin (Life Technologies Pty

Ltd., Australia). After washing, the coverslips were mounted with ProLong® Gold

(Life Technologies Pty Ltd., Australia) and secured to glass slides with three spots of

nail varnish. Morphological characteristics were identified under a fluorescent

microscope. The shapes of the fluorescent signals induced by the conditioned medium

were analysed using the integrated morphometry shape factor analysis of MetaMorph

image analysis software version 7.1.2 (Universal Imaging, Downingtown, PA, USA).

In the present study, we used a standard shape factor defined by (area/perimeter2) ×

4π for quantitative analysis of cell shape [357]. This algorithm assigns a value from 0

to 1, describing the shape of a perfect circle attains a value of 1 and a line is assigned

as 0.

RNA extraction, cDNA synthesis, and real time quantitative-PCR (RT-qPCR)

The mRNA expression of alkaline phosphatase (ALP), osteopontin (OPN) and

osteocalcin (OCN) and VEGF was measured in MSCs and OMSCs to detect the

difference before and after osteogenic differentiation. The expression of CXCL12 and

CXCR4 in MSCs was also measured after co-culture with different CM and VEGF

treatment. Total RNA was extracted from the cells with TRIzol Reagent (Ambion®,

Life Technologies Pty Ltd., Australia). Complementary DNA was synthesized from 1

μg of total RNA using DyNAmo™ cDNA Synthesis Kit (Finnzymes, Genesearch Pty

Page 83: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

63 | P a g e

Ltd., Australia) following the manufacturer’s protocol. The RT-qPCR primers (Table

1) were designed based on cDNA sequences from the NCBI Sequence database and

the primer specificity was confirmed by BLASTN searches. RT-qPCR was performed

on an ABI Prism 7300 Sequence Detection System (Applied Biosystems, Australia)

using SYBR® Green detection reagent (Applied Biosystems, Australia). In brief,

cDNA samples (2.5 μL of 1:10 diluted template for total volume of 25 μL per reaction)

were analysed for the genes of interest and for the GAPDH house keeping gene. The

cycling conditions were set as follows: 2 min at 95°C for activation followed by 45

cycles of denaturation at 95°C for 5 sec, annealing at 60°C for 10 sec, and extension

at 72°C for 15 sec. The specificity of amplification was verified by a single peak in

the melting curve. All reactions were run in triplicate for three independent

experiments. Dissociation curve analysis was performed to validate the absence of

primer dimmers and nonspecific PCR products. The mean cycle threshold (Ct) value

of each target gene was normalized against Ct value of GAPDH and the relative

expression calculated using the following formula: 2-(normalized average Cts)

×104 [358].

Table 1. Primer sequences for the gene observed in this study

Gene

name

Forward sequences Reverse sequences

ALP 5’TCAGAAGCTCAACACCAACG 5’TTGTACGTCTTGGAGAGGG

C

OCN 5’CACCGAGACACCATGAG 5’TGGAGAGGAGCAGAACTG

OPN 5’CCAAGTAAGTCCAACGAAAG 5’GGTGATGTCCTCGTCTGTA

VEGF 5’GCTGTCTTGGGTGCATTGG 5’GCAGCCTGGGACCACTTG

CXCL12 5’GCTACAGATGCCCATGCCGA

TTC

5’CCTGAATCCACTTTAGCTTC

GGGTC

CXCR4 5’CCAGAAGAAACTGAGAAGC

ATGACGG

5’CCAGACGCCAACATAGACC

ACC

GAPDH 5’ TCAGCAATGCCTCCTGCAC 5’ TCTGGGTGGCAGTGATGGC

Page 84: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

64 | P a g e

Protein extraction and Western blotting

Total protein was harvested by lysing the cells in a lysis buffer containing 20 mM

HEPES (pH 7.4), 10% glycerol, 1% Triton X-100, 2mM EGTA and a protease

inhibitor cocktail (Roche Products Pty. Ltd., Dee Why, NSW, Australia). The protein

concentration was determined by the BCA Protein Assay Kit (Thermo Fisher

Scientific, VIC, Australia). 20 µg of protein from each sample was separated by 8%

SDS-PAGE gels. The protein was then transferred onto a nitrocellulose membrane

(Pall Corporation, East Hills, NY, USA). After blocking for 1 h with blocking buffer

(1×Tris-buffered saline with 5% skim milk and 0.01% Tween-20), the membranes

were probed with primary antibodies against VEGF (1:1000 rabbit anti-human),

CXCL12 (1:1000, rabbit anti-human), CXCR4 (1:1000, rabbit anti-human) and α-

Tubulin (1:5000, rabbit anti-human) overnight at 4°C. The primary antibodies were

all obtained from Abcam (Sapphire Bioscience Pty. Ltd., Waterloo, NSW, Australia).

The membranes were washed three times for 20 min each in TBS containing 0.01%

Tween-20, and then incubated with horseradish peroxidase (HRP)-conjugated goat

anti-rabbit secondary antibody (Abcam; Sapphire Bioscience Pty. Ltd., Waterloo,

NSW, Australia) at 1:2000 dilution for 1 h at room temperature. Targeted proteins

were visualized using the SuperSignal West Femto Maximum Sensitivity Substrate

(Thermo Fisher Scientific, Australia) and exposed on x-ray film (Fujifilm, Australia).

Bands intensities were quantified by scanning densitometry and analysed using

ImageJ software [359].

Animal study and drug administration

Collagen scaffolds were fabricated from a 1mm-thick bovine Type-I collagen sheet

(Medtronic Australia Pty Ltd, North Ryde, NSW, Australia) using a hole puncher and

sterilized by soaking in 75% ethanol, followed by three rinses of PBS prior to cell

seeding. For the in vitro osteogenic differentiation, 6×104 MSCs were firstly seeded

onto collagen membrane and cultured in osteogenic medium for 2 weeks.

Page 85: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

65 | P a g e

Twelve 5-week old male SCID mice (Animal Resources Centre, Canning Vale, WA,

Australia) were used for the skull defect model [78, 321] and subcutaneous cell

implantation, respectively. For the skull defect implantation, an incision was made

longitudinally along the centre line of the shaved skull skin after anaesthesia. A 3 mm

defect was then made through the skull using a dental bur. The collagen membrane

carrying OMSCs were inserted into the defects of each mice. For the subcutaneous

cell implantation, a longitudinal incision was made in each animal along the central

line of the shaved dorsal. Each individual pocket received one collagen scaffold

carrying OMSCs. Six mice were assigned to experimental group using a complete

randomized design, which received local injection of 20 ng VEGF neutralizing

antibody in 100 uL PBS for two weeks. The other six mice in the control group

received 100 uL of PBS. All of the SCID mice were sacrificed after four weeks and

the implants were retrieved.

Micro-computed tomography (μCT) assessment

All the samples harvested were fixed in 4% PFA overnight at room temperature and

then washed in PBS. Cross-sectional scans parallel to the cell seeding surface of the

implants were performed in a micro-CT scanner (µCT40, SCANCO Medical AG,

Brüttisellen, Switzerland) at a resolution of 12 µm and three-dimensional (3D) images

of the implants were reconstructed. The calcified area inside the implant on each

scanned slice was circled out and the volume of calcification within the implants was

measured using the inbuilt scanner software package and recorded for statistical

analysis.

Histology and immunohistochemistry (IHC)

All harvested subcutaneous samples were embedded in paraffin block for slicing,

whereas the skull defect samples were firstly decalcified in 10% EDTA solution for

four weeks and then embedded in paraffin. 5 µm serial slices were cut from the

embedded samples using a microtome (Leica Microsystems GmbH, Wetzlar,

Germany). Only slices close to the centre of the samples were used for the staining.

Page 86: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

66 | P a g e

In order to observe the calcification in subcutaneous implants, sample was subjected

to von Kossa with H&E staining. The slices were dewaxed in xylene and then

hydrated in 100%, 90%, and 70% ethanol for 3 min each. After the application of 5%

silver nitrate solution (Sigma-Aldrich, Australia), the slices were exposed to light for

1 h. After rinsing in distilled water, 5% sodium thiosulphate was added to the slices

for 5 min. The slices were then washed in tap water, placed in Harris’s haematoxylin

(Sigma-Aldrich, Australia) for 2 min then rinsed in tap water before dipping 3 times

in acid ethanol. After dehydration, eosin (HD Scientific Supplies Pty Ltd., NSW,

Australia) was applied to the slices for 15 sec. The slices were then rinsed in 100%

ethanol twice and cleared in xylene before being mounted with cover slips.

IHC staining was performed for assessment of new bone formation in the skull

defects. Briefly, endogenous peroxidase activity was quenched by incubating with 3%

H2O2. After washing with PBS, all the sections were blocked with 10% swine serum

for 1 h. Sections were then incubated with optimal dilution of primary antibodies

against ALP (1:400, anti-human, Sigma-Aldrich, Australia) overnight at 4°C. Sections

were washed free of primary antibodies and then incubated with a biotinylated

secondary antibody (Dako, CA, USA) for 15 min, followed by 15 min incubation with

HRP-conjugated streptavidin-biotin complex (Dako, CA, USA). Antibody complexes

were visualised after the addition of a buffered diaminobenzidine (DAB) for 3 min.

In situ hybridisation (ISH)

To verify the origin of cells in skull defect sites, ISH was performed using a human

specific digoxigenin (DIG)-labelled oligonucleotide probe (PanPath, Budel, the

Netherlands), which detects Alu repeat sequences found in human chromosomes.

Briefly, tissue sections were dewaxed and rehydrated through a descending ethanol

series with RNase-free reagents, and then treated with proteinase K for 10 min. One

drop of the ready-to-use Alu probe solution was added onto each section and the

slices were covered with a coverslip. After heating at 95oC for 5 min, the slices were

incubated at 37°C in a humidified chamber overnight followed by post-hybridisation

Page 87: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

67 | P a g e

washing procedure. 3-Amino-9-ethylcarbazole (AEC) was then applied to visualise

the bound probe. After rinsing in distilled water, the sections were lightly

counterstained with haematoxylin, then mounted with Crystal MountTM

aqueous

mounting medium (Sigma-Aldrich, Australia).

For ISH and IHC double staining, after the donor human cells were detected using the

Alu DNA probe, which was visualised with NiCl2+DAB (staining colour is black),

the first primary antibody (VEGF) was applied and visualised with DAB (staining

colour is brown). The host mouse cells were then stained with Nuclear Fast Red

(Sigma-Aldrich, Australia). After rinsing with tap water, the sections were dehydrated

with ascending concentrations of ethanol, cleared with xylene and mounted with a

cover slip.

Statistical analysis

Statistical evaluation was performed using SPSS software (SPSS, Sigma Stat, Erkrath,

Germany). Differences between two groups were assessed by Student t-test.

Differences in the treatment outcomes from different conditions were assessed by

analysis of variance (ANOVA) with Bonferroni post-hoc test. Values of p<0.05 were

considered statistically significant.

Page 88: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

68 | P a g e

RESULTS

OMSCs express high level of VEGF

Histology images showed a close interaction between donor human OMSCs and host

mouse cells in the newly formed bone tissues from the skull defect model (Figure 14).

Furthermore, the ISH together with IHC staining revealed that the donor OMSCs

strongly expressed VEGF, giving a clue that VEGF secreted by donor OMSCs may

play a role in the host cell recruitment (Figure 14A and B).

Figure 14. Histological evaluation of a skull defect model after a 4-week implantation

of OMSCs. (A) Representative image showing host mouse cells were recruited to the bone defect

area and involved in the new bone formation with donor human cells (20×). (B) 40× magnification of

the selected area, showing the VEGF secretion from OMSCs, as well as the close interaction between

donor cells and host cells. Host mouse cells were stained with Nuclear Fast Red (pink colour, dashed

arrow pointed); Donor human cells were stained with NiCl2+DAB (black colour, arrow pointed);

VEGF was stained with DAB (brown colour, arrowhead pointed). NB: newly formed bone.

Page 89: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

69 | P a g e

Alizarin Red S staining was performed and the result showed that the mineralization

in MSCs was significantly enhanced after osteogenic differentiation (Figure 15A).

The mRNA level of ALP, OPN and OCN also showed significant increase in OMSCs

(Figure 15B, p<0.05). VEGF gene expression of OMSCs increased remarkably

(Figure 15C, p<0.05). ELISA result showed that VEGF secretion was significantly

elevated in MSCs after exposure to osteogenic medium (Figure 15D, p<0.01).

Western blotting analysis further confirmed the elevated VEGF protein level (Figure

15E, p<0.05).

Figure 15. VEGF secretion from OMSCs. (A) Alizarin Red S staining showed the

mineralization in MSCs was significantly enhanced after cultured in osteogenic medium for 2 weeks.

(B) mRNA transcripts of ALP, OCN and OPN, showed a significant increase in OMSCs (*p<0.05). (C)

VEGF gene expression of MSCs increased remarkably after osteogenic differentiation (*p<0.05). (D)

ELISA result showed that VEGF secretion significantly elevated in OMSCs (**p<0.01). (E) and (F)

Western blotting analysis showed greater VEGF expression in OMSCs in the protein level, as indicated

by the significantly higher band density (*p<0.05).

Page 90: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

70 | P a g e

VEGF secreted by OMSCs induced the migration of MSCs

After 8 h incubation, the migration of MSCs through the transwell membrane was

significantly enhanced in the co-culture with OMSCs compared with the serum-free

medium or the co-culture with MSCs (Figure 16C, D and I, p<0.01). However, the

number of migrated MSCs dropped dramatically when the OMSCs were treated with

VEGF neutralizing antibody (Figure 16D and J, p<0.01). The increased motility of

MSCs toward OMSCs was further confirmed using the xCELLigence system. The

cell index of MSCs in CMOMSC

was elevated throughout the experimental period, but

dropped down to the level as that in the CMMSC

when VEGF neutralizing antibody

was applied (Figure 16K, p<0.05). The data demonstrated that VEGF secreted by

OMSCs serves as a chemoattractant to MSCs.

Morphological changes of MSCs cultured in CMOMSC

As described above, enhanced migration of MSCs was observed in the indirect co-

culture with OMSCs, indicating a more mobile MSC phenotype was induced. We

then further investigated the effect of CMOMSC

on the morphological and cytoskeletal

changes in MSCs. Figure 17 shows MSCs cultured in CMOMSC

underwent remarkable

morphological alterations compared with those cultured in CMMSC

over the same

period of time. Actin filaments organized along the length of the cell, making the

general appearance of cell as an elongated rod shape (Figure 17B). However, the

application of VEGF neutralizing antibody in CMOMSC

led to a reversed effect (Figure

17C). Cell shape factor changed from around 0.5 to 0.1, indicating a transformation

from a more regular and round shape to an irregular and elongated cell shape when

MSCs were grown in the CMOMSC

instead of CM

MSC. Additionally, when VEGF

neutralizing antibody was added into the CMOMSC

, the cells tended to regain a higher

shape factor, indicative of the importance of VEGF in maintaining the migratory

phenotype (Figure 17D, p<0.05).

Page 91: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

71 | P a g e

Figure 16. VEGF neutralizing antibody interfered with the migration of MSCs

towards OMSCs. (A)-(D) and (I) The migration capacity of MSCs towards OMSCs was

significantly superior to the control groups (SF, serum-free medium; MSC, undifferentiated

mesenchymal stromal cells; OMSC, osteogenically differentiated mesenchymal stromal cells;

**p<0.01). (A)-(D) and (J) The number of migrated MSCs decreased significantly with the addition of

20 ng/mL VEGF neutralizing antibody (*p<0.05). (K) The increased motility of MSCs toward OMSCs

was further confirmed by the xCELLigence system (*p<0.05).

Page 92: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

72 | P a g e

Figure 17. Morphological and cytoskeletal changes in MSCs cultured in CMOMSC

with or without the addition of VEGF neutralizing antibody. (A)-(C) MSCs cultured in

CMOMSC showed elongated actin filaments. The application of VEGF neutralizing antibody in CMOMSC

led to a reversed effect. (D) The shape factor of MSCs cultured in CMOMSC was significantly lower than

that in CMMSC and CMOMSC + Abvegf (*p<0.05).

Page 93: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

73 | P a g e

CMOMSC

increased the CXCL12/CXCR4 expression of MSCs

We then further tested the effect of VEGF secreted by OMSCs on the expression

pattern of CXCL12/CXCR4, which is believed to be the most important axis that

contributes to MSCs homing to areas of tissue damage. When MSCs were cultured in

CMOMSC

, the CXCL12/CXCR4 mRNA expression showed a significant elevation

(Figure 18A and B, p<0.05) compared to those cultured in CMMSC

. Furthermore, the

expression of CXCL12/CXCR4 decreased when VEGF neutralizing antibody was

added into CMOMSC

(Figure 18A and B, p<0.05). The change of CXCL12/CXCR4

expression was further confirmed by the Western blotting results (Figure 18C-E,

p<0.05).

Figure 18. CMOMSC

increased the CXCL12/CXCR4 expression of MSCs. (A) and (B)

mRNA levels of CXCL12 and CXCR4 were significantly elevated in MSCs cultured in CMOMSC

(*p<0.05). However, the expression of CXCL12/CXCR4 decreased when VEGF neutralizing antibody

was applied (*p<0.05). (C)-(E) The elevated expression of CXCL12/CXCR4 was further confirmed by

the Western blotting analysis on the protein level, as indicated by the significantly higher band density

(*p<0.05). CMMSC

, conditioned medium derived from undifferentiated mesenchymal stromal cells;

CMOMSC, conditioned medium derived from osteogenically differentiated mesenchymal stromal cells;

Abvegf, VEGF neutralizing antibody.

Page 94: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

74 | P a g e

CXCL12/CXCR4 expression in MSCs was stimulated by VEGF

The effect of VEGF on the expression of CXCL12/CXCR4 in MSCs was further

investigated by culturing the MSCs in medium supplemented with increasing

concentration of VEGF. The results showed that the CXCL12/CXCR4 gene

expression in MSCs enhanced significantly when stimulated by VEGF in a dose

dependant manner (Figure 19A and B, p<0.05). Western blotting analysis showed a

similar trend on the protein level (Figure 19C-E, p<0.05).

Figure 19. CXCL12/CXCR4 expression of MSCs stimulated by VEGF. (A) and (B)

mRNA levels of CXCL12 and CXCR4 in MSCs treated with different dosage of VEGF, showing a

steady increase in a dose dependent manner (*p<0.05). (C)-(E) The elevated expression of

CXCL12/CXCR4 in MSCs stimulated with increasing dosage of VEGF was further confirmed by the

Western blotting analysis on the protein level, as indicated by the significantly higher band density

(*p<0.05).

Page 95: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

75 | P a g e

Inhibition of VEGF abrogated the effect of OMSCs on the recruitment of host cells

To determine whether VEGF secreted by OMSCs plays a role in host cell recruitment

in vivo, donor cells were implanted into skull defects of SCID mice with or without

the local administration of VEGF neutralizing antibody. The von Kossa with H&E

staining revealed the mineralization found in the subcutaneous implants carrying

OMSCs was suppressed when the VEGF neutralizing antibody was applied (Figure

20A and B). For the skull defect samples, µCT scanning and 3D reconstruction

confirmed that VEGF neutralizing antibody significantly inhibited the bone healing

induced by the implantation of the OMSCs. (Figure 20E and F). IHC staining against

ALP revealed less new bone tissues in the skull defects after the local application of

VEGF neutralizing antibody (Figure 20C and D). The results suggested that VEGF

was involved in the accelerated bone healing induced by OMSC implantation. ISH

against human Alu sequences revealed that host mouse cells (Alu negative) were

recruited to the skull defect area and actively involved in the bone regeneration

process together with the donor human OMSCs (Figure 20G). On the contrary, fewer

host cells were found in the implants when VEGF was neutralized (Figure 20H).

Page 96: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

76 | P a g e

Figure 20. Blocking VEGF with neutralizing antibody resulted in a significant

decrease of MSC recruitment and new bone formation. (A) von Kossa with H&E staining

showed mineralization in the subcutaneous implants carrying OMSCs. (B) von Kossa with H&E

staining detected no mineralization when the VEGF neutralizing antibody was applied. (C)

Immunohistochemical staining against ALP revealed newly formed bone in skull defect implanted with

collagen scaffold carrying OMSCs. (D) Less new bone formation was detected in the skull defects after

the local application of VEGF neutralizing antibody. (E) and (F) 3D images of skull defects implanted

with collagen scaffold carrying OMSC, with or without the application of VEGF neutralizing antibody.

(G) ISH staining revealed that host mouse cells (stained in purple) were recruited to the skull defect

area and involved in the bone regeneration together with the donor human OMSCs (stained in red). (H)

Fewer host cells were recruited to the implants when VEGF was neutralized.

Page 97: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

77 | P a g e

DISCUSSION

Cell implantation is currently regarded as an important and promising regime for

repairing and restoring the biological functions of bone defects [307, 308]. Bone

marrow-derived MSCs are promising candidates for such cell-based therapy, due to

their immune tolerance, self-renewal capacity and ease of use [360-362]. Chemotactic

migration of host mesenchymal progenitor cells (MPCs) from bone marrow or other

tissues have been identified as a crucial event during various physiological and

pathological processes [351, 353, 354]. Increasing evidence indicates that MSCs

communicate with other cells in vivo and appear to “home” to areas of defect and

injury in response to cellular signals. However, even though the therapeutic potential

of MSCs is an area of great excitement and promise, the factors that trigger MSCs’

response and the underlying mechanisms that guide the host MSCs migrate to the

bone defect sites are still poorly understood. In this study we have demonstrated that

VEGF secreted by OMSCs led to the altered expression pattern of CXCL12/CXCR4

in MSCs, and we have also provided some insights into the communication between

host cells and donor cells during osteogenesis.

MSCs secrete numerous cytokines including VEGF, bFGF, IGF-1 and HGF [363].

Among all these, VEGF is the key regulator in a cascade of molecular and cellular

events that ultimately lead to the development of the vascular system [364]. VEGF is

not only a critical mediator in physiological angiogenesis, but also a vital factor in

skeletal growth [365]. Previous studies suggested that VEGF plays an important role

in MSCs migration [366-368], as well as in the recruitment and proliferation of

endothelial cells (ECs) [369]. The data from our study showed that the VEGF

secretion increased dramatically in bone marrow-derived MSCs after osteogenic

differentiation in vitro. Similar results have been reported previously that VEGF is

expressed in osteoblast-like cells in a differentiation-dependent manner, indicating

that VEGF plays a positive role in the regulation of osteoblast activity [199]. A recent

study has also demonstrated that Osterix (Osx), an essential osteoblast-specific

transcription factor required for osteoblast differentiation, directly targets VEGF

expression [201], suggesting a close link between osteogenesis and angiogenesis. The

enhanced secretion of VEGF from OMSCs was further confirmed by the ISH and

Page 98: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

78 | P a g e

IHC staining, which showed a strong expression of VEGF in OMSCs correlated with

greater recruitment of host MSCs.

We then demonstrated the positive effect of VEGF on the motility of MSCs in vitro

using transwell migration assay and real-time xCELLigence system. The results

suggested the secreted factors derived from OMSCs were a potent chemoattractant for

MSCs. The enhanced motility of MSCs was correlated with the changes in actin

organization and distribution from azimuthal symmetry around the cell rim to

concentration at a particular region [370]. In addition to morphological changes,

MSCs underwent specific alterations in gene expression patterns in response to the

exterior stimuli. Exposure to CMOMSC

enhanced migration of MSCs and this was

accompanied by an increased expression pattern of CXCL12/CXCR4. In agreement

with the gene expression profile, CXCL12/CXCR4 protein levels also increased in

MSCs upon exposure to CMOMSC

. However, the addition of VEGF neutralizing

antibody abrogated the effect of OMSCs on the actin filament arrangement as well as

the CXCL12/CXCR4 expression of MSCs. We further confirmed a dose-dependent

chemoattractive effect of VEGF on MSCs. VEGF stimulation remarkably increased

the expression of CXCL12/CXCR4, which is the best known axis that contributes to

cell migration [352]. Additionally, the expression of CXCL12/CXCR4 in MSCs was

down regulated when VEGF neutralizing antibody was applied, suggesting the

secretion of VEGF from OMSCs played a crucial role in inducing the MSCs

recruitment.

The interaction between donor cells and host cells is an emerging area of research,

and it is important to understand the mechanisms that govern this interaction. Our data

has shown that there was differential regulation of CXCL12/CXCR4 in MSCs

exposed to different microenvironments and that these changes influenced chemotaxis,

an important function of MSCs. Based on the previous finding, our in vivo study

further confirmed that OMSCs played a significant role in the host cells recruitment.

It is very interesting to find that the transplanted OMSCs were involved in the new

bone formation. Furthermore, the function of OMSCs in host cell recruitment was

Page 99: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

79 | P a g e

supported by the elevated secretion of VEGF, which then led to the markedly

increased expression of CXCL12/CXCR4 and changes of morphology in host MSCs

(Figure 21). CXCL12 may signal through its receptor, CXCR4, and function in an

autocrine manner to support chemotaxis of host cells towards the transplanted donor

cells in the bone defect sites. The newly formed blood vessels, donor cell- and host

cell-derived osteoblasts may work together to restore the damaged bone tissue.

Further studies are needed to elucidate the full repertoire of CXCL12/CXCR4

signaling in MSCs and to identify other pathways that are critical to the

microenvironment-specific MSC biology. This will be crucial to the understanding of

clinically important processes, such as cell transplantation, bone marrow engraftment

and wound healing.

Figure 21. Schematic of the proposed mechanism by which donor OMSCs initiate a

paracrine signaling cascade that results in altered phenotype and enhanced motility of

host MSCs in a skull defect model. The interaction between OMSCs and MSCs is centrally

regulated by VEGF secreted by OMSCs, and involves the participation of the CXCL12/CXCR4 axis,

which may accelerate the host cell recruitment process.

Page 100: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

80 | P a g e

CONCLUSION

We demonstrated the VEGF secreted by OMSCs plays a significant role in the

expression pattern of CXCL12/CXCR4 of undifferentiated bone marrow-derived

MSCs. The involvement of CXCL12/CXCR4 in the VEGF-mediated host cell

recruitment towards skull defect was confirmed for the first time. The present study

offers insights into the donor/host cell interaction and is of clinical significance for the

application of cell-based therapy for bone repair and regeneration.

Page 101: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

81 | P a g e

REPORT THREE

Involvement of VEGF/PDGFR and the PI3K/Akt signaling pathway in

OMSCs-induced MSCs migration

Yinghong Zhou, Ross Crawford, Yin Xiao

Suggested Statement of Contribution of Co-Authors for Thesis by

Published Papers:

Contributors Statement of contribution*

Yinghong Zhou Involved in the conception and design of the project, performed

laboratory experiments, data analysis and interpretation. Wrote

the manuscript.

Ross Crawford Assisted in collection of clinical samples and reviewed the

manuscript.

Yin Xiao Involved in the conception and design of the project, and

reviewed the manuscript.

Principal Supervisor Confirmation

I have sighted email or other correspondence from all co-authors confirming

their certifying authorship.

Name Signature Date

Page 102: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

82 | P a g e

ABSTRACT

Background: Although it has been shown that mesenchymal stromal cells (MSCs)

are attracted to vascular endothelial growth factor (VEGF) secreted by osteogenically

differentiated MSCs (OMSCs) in our previous study, it is still not known that how

VEGF signals through MSCs which are expected to have low levels of VEGF

receptors (VEGFRs) and what downstream pathways are involved in the recruitment

of MSCs.

Methods: MSCs were stimulated with different concentrations of VEGF, conditioned

medium derived from MSCs (CMMSC

) or OMSCs (CMOMSC

) to establish the receptors’

activation profile. The role of platelet-derived growth factor receptors (PDGFRs) was

further investigated using phospho-receptor tyrosine kinase (RTK) array, Western

blotting analysis and in vitro migration assay. The involvement of the PI3K/Akt

signaling pathway in OMSCs-induced MSC migration was monitored by relevant

phosphorylated antibodies and inhibitors.

Results: We found that VEGF secreted by OMSCs was capable of inducing migration

of MSCs which are VEGFR deficient, and this induction was mediated through a

molecular crosstalk of PDGFRs and the PI3K/Akt signaling kinase. The VEGF-

induced MSC migration paralleled with the enhanced expression of PDGFRs and the

activation of the PI3K/Akt kinase. Blocking of PDGFRs activity impaired VEGF-

induced MSC migration as well as PI3K/Akt kinase activation. Moreover, a PI3K/Akt

specific inhibitor, LY294002, resulted in the suppression of VEGF-induced MSC

migration and dephosphorylation of downstream effectors of the PI3K/Akt signaling

pathway, including Akt, mTOR, p70S6K

and PAK1, further supporting the role of the

PI3K/Akt pathway in the MSC recruitment.

Conclusions: The present study established a mechanistic link among VEGF,

PDGFRs and PI3K/Akt in the regulation of MSC recruitment that eventually may be

beneficial to the bone repair and regeneration.

Page 103: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

83 | P a g e

INTRODUCTION

The molecular interactions between donor and host cells are indispensable in the

process of host mesenchymal stromal cells (MSCs) recruitment for tissue repair and

regeneration. Guidance of cell migration depends critically on subcellularly localised

perception and transduction of various signals. As has been widely reported, vascular

endothelial growth factor (VEGF) proved to be chemoattractive for primary human

osteoblasts [202, 203]. This provides the first evidence that VEGF may also be

involved in local recruitment of osteoprogenitor cells in the course of endochondral

bone formation or remodeling. VEGF exerts its effects on vascular cells by binding to

three structurally related VEGF receptor (VEGFR) tyrosine kinases, VEGFR-1,

VEGFR-2 and VEGFR-3, each forms homodimers on ligand binding [206]. VEGF

and platelet-derived growth factor (PDGF) are closely related members, as evidenced

by sequence analysis which predicts that VEGF and PDGF evolved from a common

ancestor [207]. VEGFRs and PDGF receptors (PDGFRs) are also structurally related,

characterised by extracellular immunoglobulin-like domains with an intracellular

tyrosine kinase domain interrupted by a non-catalytic region [371]. Bone marrow-

derived MSCs, which can differentiate into vascular cells, may be recruited by VEGF

during the process of angiogenesis and osteogenesis. However, several studies have

reported that there is limited expression of VEGFR in MSCs [200, 372], leading to the

speculation that VEGF may signal through PDGFRs and regulate the migration of

MSCs [210].

Our previous study (REPORT TWO) has demonstrated that VEGF secreted from

osteogenically differentiated mesenchymal stromal cells (OMSCs) played a

significant role in the recruitment of host cells by regulating the expression pattern of

CXCL12/CXCR4, an important axis for cell migration. The present study aimed to

identify the downstream signaling pathways associated with the interaction between

OMSCs and MSCs. Recent studies have reported that binding of CXCL12 to CXCR4

may lead to stimulation of signaling cascade that involves activation of multiple

targets, including Erk1/2, p38 and Akt [273-276]. Akt, also known as protein kinase B

(PKB), is one of the best characterised targets of phosphoinositide 3-kinases (PI3K)

signaling pathway, which regulates cell survival, cytoskeletal rearrangements and cell

Page 104: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

84 | P a g e

invasion [252, 253]. Accumulating data suggest that major downstream effectors of

the PI3K/Akt signaling pathway, mTOR, p70 S6 kinase (p70S6K

) and p21-activated

kinase 1 (PAK1), are associated with changes in actin cytoskeleton organization, cell

shape and adhesion dynamics, and play critical roles in cell motility regulation [281,

282, 285-287].

Here in the present study, we found that OMSCs stimulated the migration of MSCs by

activating PDGFRs. Furthermore, our findings demonstrated VEGF/PDGFR mediated

the activation of the PI3K/Akt signaling cascade, which led to the phosphorylation of

mTOR, p70S6K

and PAK1, and was significantly associated with the cell migration

induced by OMSCs.

MATERIALS AND METHODS

Cells and Cell culture

Human bone marrow samples were collected from patients (n=3, 1 female and 2

males) undergoing elective knee replacement surgery in the Department of

Orthopaedics at Prince Charles Hospital, Brisbane, Queensland, Australia after

obtaining informed consent from each donor and ethics approval from the Ethics

Committee of Queensland University of Technology. Bone marrow-derived MSCs for

this study were isolated by density gradient centrifugation over Lymphoprep (Axis-

shield PoC AS, Oslo, Norway) according to the manufacturer’s protocol. The cells

were then seeded in culture flasks supplemented with low glucose Dulbecco’s

Modified Eagle Medium (DMEM; Gibco®, Life Technologies Pty Ltd., Australia)

containing 10% (v/v) fetal bovine serum (FBS; In Vitro Technologies, Australia) and

1% (v/v) penicillin/streptomycin (P/S; Gibco®, Life Technologies Pty Ltd., Australia)

at 37°C, 5% CO2. The medium was changed twice weekly to wash out all non-

adherent cells. Adherent bone marrow-derived MSCs were then passaged once grown

to 70%-80% confluence. Cell characterisation for MSCs has been carried out in our

previous study [318, 355]. Human umbilical vein endothelial cells (HUVECs;

Clonetics™, Lonza Australia Pty Ltd., Mt Waverley, VIC, Australia) were used as

Page 105: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

85 | P a g e

positive control when required. HUVECs were routinely cultured in EGM™-2

BulletKits™ (Lonza Australia Pty Ltd., Mt Waverley, VIC, Australia) composed of

endothelial cell basal medium-2 (EBM-2) supplemented with 2% FBS,

gentamicin/amphotericin-B, VEGF, human epidermal growth factor (hFGF)-B,

insulin-like growth factor-1 (IGF-1), hEGF, ascorbic acid, hydrocortisone, and

heparin, as described by the manufacturer (Lonza Australia Pty Ltd., Mt Waverley,

VIC, Australia).

Osteogenic differentiation of bone marrow-derived MSCs

Isolated MSCs were subjected to osteogenic differentiation for 2 weeks in osteogenic

medium (DMEM supplemented with 10% FBS, 1% penicillin/streptomycin, 10 mM

β-glycerophosphate, 50 µM ascorbic acid and 100 nM dexamethasone; Sigma Aldrich,

Australia) for further experiments.

Preparation of conditioned medium (CM)

CM was obtained from 1×107

MSCs (CMMSC

) or OMSCs (CMOMSC

). The cells were

washed twice with PBS, and incubated with 10 mL of serum-free DMEM at 37 oC for

48 h. The CM was then collected, subjected to centrifugation at speed of 1000 g for 5

min at 4oC and filtration through a 0.2 µm low protein binding filter (Millipore

Corporation, Billerica, MA, USA) to remove cell debris. The supernatants were stored

at -80oC for subsequent experiments.

Stimulation of MSCs

In order to investigate the effect of VEGF on the expression pattern of VEGFRs and

PDGFRs, cell culture medium supplemented with 20, 40, 80 ng VEGF (R&D

Systems Inc., Australia) per mL was applied to HUVECs and MSCs. The cells were

harvested and subjected to real time quantitative PCR (RT-qPCR) analysis after 24 h.

MSCs were also stimulated with serum-free medium, CMMSC

, CMOMSC

with or

Page 106: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

86 | P a g e

without the supplementation of 20 ng/mL VEGF neutralizing antibody (R&D Systems

Inc., Australia) to further investigate the expression pattern of various receptors.

For the functional tests, MSCs were pre-treated with 5 µM PDGFR tyrosine kinase

inhibitor (Merck Pty Ltd., Australia), 10 µM LY294002 (Cell Signaling Technology,

Inc. Danvers, MA, USA) or DMSO as control 30 min prior to the stimulation of

CMMSC

and CMOMSC

. MSCs were harvested for RNA and protein at different time

points (e.g. 15 min, 30 min and 60 min) according to subsequent experiments.

In vitro migration assay using transwell chambers

MSCs migration was monitored using 6-well transwell cell culture Boyden chambers

with 23.1 mm effective diameter polyethylene terephthalate (PET) membrane

containing 8 µm pores (Becton-Dickinson Labware, Franklin Lakes, NJ, USA). 5×104

MSCs were firstly seeded on to each transwell chamber for the initial attachment.

After serum-starved overnight, the transwell chambers were assembled onto the 6-

well companion plates supplemented with CM from MSCs or OMSCs. To attenuate

the PDGFR in MSCs, 5 µM PDGFR tyrosine kinase inhibitor was applied. Non-

specific effects of the inhibitor were ruled out by repeating the migration assay in the

presence of an unrelated antibody against an intracellular antigen IgG. Cell migration

was allowed to proceed for 8 h. Cells that had migrated to the other side of the

transwell membrane were fixed, and stained with crystal violet. Images were taken

with an inverted microscope (Eclipse TS100, Nikon Australia Pty Ltd.). The

migration was quantified by counting the number of cells that migrated through the

pores in six randomly selected fields on each transwell membrane, and values from

three independent experiments, with triplicate samples in each experiment, were

averaged and shown as mean ± SD.

Page 107: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

87 | P a g e

Phospho-receptor tyrosine kinase (RTK) array

A human phospho-receptor tyrosine kinase (RTK) array kit (R&D Systems Inc.,

Australia) was used to simultaneously detect the relative tyrosine phosphorylation

levels of 49 different RTKs in the untreated or CM-treated MSC lysates. Each array

contains duplicate validated controls and captures antibodies for specific RTKs.

MSCs were serum-starved for 12 h and then stimulated with CMMSC

or CMOMSC

for

15 min at 37°C in a humidified atmosphere with 5% CO2, then immediately placed on

ice, washed twice with chilled PBS, and harvested using the lysis buffer that comes

with the RTK array kit. Total protein concentration was quantitated using the BCA

Protein Assay Kit (Thermo Fisher Scientific, VIC, Australia). RTK array analysis was

performed according to the manufacturer’s protocol. In brief, the array membranes

were blocked, and then incubated with 250 µg MSC lysate overnight at 4°C. After

washing, the membranes were incubated with anti-phospho-tyrosine-HRP for 2 h at

room temperature, then washed again, and developed with detection reagent. RTK

spots were visualized on x-ray film (Fujifilm, Australia).

RNA extraction, cDNA synthesis, and real time quantitative-PCR (RT-qPCR)

Total RNA was extracted from the cells with TRIzol® Reagent (Ambion

®, Life

Technologies Pty Ltd., Australia). Complementary DNA was synthesized from 1 μg

of total RNA using DyNAmo™ cDNA Synthesis Kit (Finnzymes, Genesearch Pty

Ltd., Australia) following the manufacturer’s protocol. The RT-qPCR primers (Table

2) were designed based on cDNA sequences from the NCBI Sequence database and

the primer specificity was confirmed by BLASTN searches. RT-qPCR was performed

on an ABI Prism 7300 Sequence Detection System (Applied Biosystems, Australia)

using SYBR® Green detection reagent Applied Biosystems, Australia). In brief,

cDNA samples (2.5 μL of 1:10 diluted template for total volume of 25 μL per reaction)

were analysed for the genes of interest and for the GAPDH house keeping gene. All

reactions were run in triplicate for three independent experiments. Dissociation curve

analysis was performed to validate the absence of primer dimmers and nonspecific

PCR products. The mean cycle threshold (Ct) value of each target gene was

Page 108: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

88 | P a g e

normalized against Ct value of GAPDH and the relative expression calculated using

the following formula: 2-(normalized average Cts)

×104 [358].

Table 2. Primer sequences for the gene observed in this study

Gene

name

Forward sequences Reverse sequences

VEGFR-1 5’CCCTTATGATGCCAGCAAGT

G

5’CCAAAAGCCCCTCTTCCAA

VEGFR-2 5’CAAACGCTGACATGTACGG

TCTA

5’CCAACTGCCAATACCAGTG

GAT

VEGFR-3 5’GTCAGGGGAGATCATCGGG

AC

5’GGAGCTGGTGGTGAATGTG

CCC

PDGFR-α 5’GATAGCTTCCTGAGCCACCA

C

5’CATAGAGTGATCTCTGGAT

GTCGG

PDGFR-β 5’GGAGACCCGGTATGTGTCA

GAGC

5’CCAGCACTCGGACAGGGAC

ATTG

GAPDH 5’ TCAGCAATGCCTCCTGCAC 5’TCTGGGTGGCAGTGATGGC

Protein extraction and Western blotting

Total protein was harvested by lysing the cells in a lysis buffer containing 20 mM

HEPES (pH 7.4), 10% glycerol, 1% Triton X-100, 2mM EGTA and a

phosphatase/protease inhibitor cocktail (Roche Products Pty. Ltd., Dee Why, NSW,

Australia). The protein concentration was determined by the BCA Protein Assay Kit

(Thermo Fisher Scientific, VIC, Australia). 15 µg of protein from each sample was

separated by 8% SDS-PAGE gels, and then the separated protein was transferred to

nitrocellulose membrane (Pall Corporation, East Hills, NY, USA). After blocking for

1 h with Odyssey® blocking buffer (Millennium Science, Australia), the membranes

were incubated with the primary antibodies against PDGFR-α (1:1000, rabbit anti-

human), PDGFR-β (1:1000, rabbit anti-human), Akt (1:2000, mouse anti-human),

phospho-Akt (Ser473) (1:2000, rabbit anti-human), Erk1/2 (1:2000, mouse anti-

human), phospho-Erk1/2 (1:2000, rabbit anti-human), mTOR (1:1000, mouse anti-

Page 109: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

89 | P a g e

human), phospho-mTOR (1:1000, rabbit anti-human), p70S6K

(1:1000, rabbit anti-

human), phospho-p70S6K

(Thr389) (1:1000, rabbit anti-human), PAK1 (1:1000, rabbit

anti-human), phospho-PAK1 (Ser144)/PAK2 (Ser141) (1:1000, rabbit anti-human),

and α-Tubulin (1:4000, rabbit anti-human) overnight at 4oC. The primary antibodies

were all obtained from Cell Signaling (Cell Signaling Technology, Inc. Danvers, MA,

USA). The membranes were washed three times for 20 min each in TBS containing

0.01% Tween-20, and then incubated with corresponding fluorescent secondary

antibodies (Cell Signaling Technology, Inc. Danvers, MA, USA) at 1:2000 dilution

for 1 h at room temperature. Targeted proteins were visualized using the Odyssey®

infrared imaging system. Band intensities were quantified by scanning densitometry

and analysed using ImageJ software [359].

Statistical analysis

Statistical evaluation was performed using SPSS software (SPSS Inc., Chicago, Il,

USA). Values are expressed as mean ± standard deviation (SD). One-way analysis of

variance followed by Student’s t-test was used to determine statistically significant

differences between experimental and control samples. Significant difference was

considered at p<0.05.

RESULTS

Expression profile of VEGFR and PDGFR in MSCs

RT-qPCR analysis was performed using total RNA isolated from MSCs, with human

umbilical vein endothelial cells (HUVECs) as positive control. Only a very low level

of VEGFR-1, VEGFR-2, or VEGFR-3 mRNA transcripts were determined in MSCs

compared to HUVECs, with or without the stimulation of different concentration of

VEGF, reflecting the lack of VEGFRs in MSCs (Figure 22). On the contrary, both

VEGFR-1 and VEGFR-2 were readily detected in HUVECs, showing a steady

increase after exposure to VEGF (Figure 22). Interestingly, the treatment of MSCs

with different concentration of VEGF under serum-free condition significantly

augmented the expression of PDGFR-α and PDGFR-β in MSCs (Figure 22, p<0.05).

Page 110: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

90 | P a g e

Figure 22. The expression of VEGFR and PDGFR mRNA transcripts was examined

by RT-qPCR analysis, using HUVECs as positive control. H: HUVECs; B: bone marrow-

derived mesenchymal stromal cells; SF: serum-free medium; BCM: conditioned medium from bone

marrow-derived MSCs; OCM: conditioned medium from osteogenically differentiated bone marrow-

derived MSCs; 20v: 20 ng/mL VEGF; 40v: 40 ng/mL VEGF; 80v: 80 ng/mL VEGF.

Page 111: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

91 | P a g e

Furthermore, the co-culture with CM derived from OMSCs enhanced the expression

of PDGFRs, but failed to reveal an increase in the levels of VEGFRs. Importantly, the

application of 20 ng/mL VEGF neutralizing antibody impaired the PDGFRs

expression in MSCs induced by CMOMSC

. These data indicated that PDGFR-α and

PDGFR-β may be the key players in VEGF dependent or CMOMSC

-induced migration

of MSCs, whereas VEGFRs, although required for VEGF-mediated endothelial cells

(ECs) migration [373], are unlikely to be involved.

CMOMSC

-induced PDGFR-α and PDGFR-β tyrosine phosphorylation in MSCs

Having demonstrated the enhanced expression profile of PDGFRs in MSCs after

stimulated with CMOMSC

, we next examined whether CMOMSC

could lead to PDGFR

tyrosine autophosphorylation by using a human phospho-RTK array containing 49

different specific RTK antibodies. RTK screening of CMOMSC

treated MSCs revealed

distinct PDGFR-α and PDGFR-β tyrosine phosphorylation (Figure 23C). In

comparison, the cell lysate from serum-free medium or CMMSC

treated MSCs showed

a barely detectable level of tyrosine phosphorylation (Figure 23A and B). The

neutralization of VEGF was accompanied by a significant decrease in CMOMSC

-

induced PDGFRs activation (Figure 23D). Interestingly, no VEGFR tyrosine

phosphorylation was detected, further validating that the MSCs biological activity

stimulated by CMOMSC

was not mediated through VEGFRs.

Western blotting analysis also demonstrated that, compared to CMMSC

, CM

OMSC

enhanced PDGFR-α and PDGFR-β expression of MSCs in protein level (Figure 24A).

The supplementation of VEGF neutralizing antibody led to a substantial decrease of

PDGFR expression in MSCs (Figure 24A-C, p<0.05), indicating that CMOMSC

-

induced MSCs activity may be dependent on a PDGFR-mediated mechanism.

Page 112: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

92 | P a g e

Figure 23. CMOMSC

-induced tyrosine phosphorylation of PDGFR-α and PDGFR-β in

MSCs. Human phospho-RTK array was used to examine RTK phosphorylation levels in MSCs lysate

samples. RTK array analysis of (A) MSCs cultured in serum-free medium without supplementation of

exogenous growth factor; (B) MSCs cultured in CMMSC

; (C) MSCs stimulated with CMOMSC

; (D)

MSCs cultured in CMOMSC supplemented with VEGF neutralizing antibody.

Page 113: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

93 | P a g e

Figure 24. The levels of protein expression of PDGFR-α and PDGFR-β in MSCs in

response to different stimulations. (A) Western blotting evaluation of PDGFR-α and PDGFR-β

expressions of MSCs in protein level after stimulation with CMMSC and CMOMSC with or without the

addition of VEGF neutralizing antibody. (B) and (C) Quantitative measurement of the protein band

density of PDGFR-α and PDGFR-β. The expressions of PDGFR-α and PDGFR-β elevated when MSCs

were stimulated with CMOMSC. The addition of VEGF neutralizing antibody in CMOMSC led to reduced

levels of protein expression of both PDGFR-α and PDGFR-β (*p<0.05). CMMSC, conditioned medium

derived from undifferentiated mesenchymal stromal cells; CMOMSC, conditioned medium derived from

osteogenically differentiated mesenchymal stromal cells; Abvegf, VEGF neutralizing antibody.

Role of PDGFRs in CMOMSC

-induced MSC migration

To examine whether PDGFRs participate in CMOMSC

-induced MSC migration which

has been proven in the previous study (REPORT TWO), the activity of PDGFRs was

blocked by treating the MSCs with PDGFR tyrosine kinase inhibitor for 8 h. In vitro

transwell migration assay revealed that pre-treatment of PDGFR tyrosine kinase

Page 114: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

94 | P a g e

inhibitor mediated inhibition of PDGFRs action, which significantly impaired

CMOMSC

-induced MSC migration, resulting in substantial reduction of migrated cells

(Figure 25E, F and G, p<0.05). On the other hand, pre-treatment with either VEGFR-

1 or VEGFR-2 inhibitor did not alter CMOMSC

-induced MSC migration (p>0.05; data

not shown).

Figure 25. The involvement of PDGFR in MSC migration induced by CMOMSC

. (A)-(C)

Schematics representation of in vitro migration assay settings. The bottom chambers were

supplemented with (A) CMMSC, (B) CMOMSC and (C) CMOMSC with PDGFR tyrosine kinase inhibitor.

(D)-(F) Representative images of MSC migration induced by (D) CMMSC, (E) CMOMSC and (F) CMOMSC

with PDGFR tyrosine kinase inhibitor. (G) The migration capacity of MSCs was significantly

stimulated by CMOMSC compared to CMMSC. PDGFR tyrosine kinase inhibitor was applied to attenuate

the PDGFR bioactivity and the number of migrated MSCs decreased dramatically (*p<0.05). CMMSC,

conditioned medium derived from undifferentiated mesenchymal stromal cells; CMOMSC, conditioned

medium derived from osteogenically differentiated mesenchymal stromal cells; PDGFRi, PDGFR

tyrosine kinase inhibitor.

Page 115: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

95 | P a g e

Activation of the PI3K/Akt signaling is required for CMOMSC

-induced MSC

migration

To determine the signaling pathways involved in the CMOMSC

-induced MSC

migration, we then examined the effect of PDGFR tyrosine kinase inhibitor on the

phosphorylation of Akt and Erk1/2 in the co-culture system. MSCs were stimulated

with CMMSC

and CMOMSC

with or without PDGFR tyrosine kinase inhibitor after

serum-starved overnight. Following treatment, the phospho- and total levels of Akt

and Erk1/2 were determined by Western blotting analysis. As shown in Figure 26,

MSCs displayed a very low basal Akt phosphorylation after exposure to CMMSC

.

After stimulation with CMOMSC

, a significant increase of Akt phosphorylation was

produced, but the addition of PDGFR tyrosine kinase inhibitor resulted in a decreased

phosphorylation. However, there was no major difference in the expression profile of

phospho-Erk1/2.

In order to further confirm the involvement of the PI3K/Akt signaling pathway in

CMOMSC

-induced MSC migration, MSCs were pre-treated with 10 µΜ LY294002 for

30 min prior to their exposure to CMOMSC

for different time points (e.g. 15 min, 30

min, and 60 min). As shown in Figure 27, CMOMSC

induced a dramatic and rapid

elevation of phospho-Akt, reaching a maximum within 30 min of exposure followed

by a gradual decrease observed 60 min after CMOMSC

stimulation. No significant

change in total Akt expression was observed over the course of the experiment. The

interference with the PI3K/Akt signaling by LY294002 abolished the signal

transduction (Figure 27A and B, p<0.05). Furthermore, the inhibition of the

PI3K/Akt signaling pathway was associated with significantly reduced migration of

MSCs induced by CMOMSC

as shown in the in vitro migration assay (Figure 27C,

p<0.05).

Page 116: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

96 | P a g e

Figure 26. Akt and Erk1/2 phosphorylation status in MSCs in response to various

stimulations. (A) Western blotting evaluation of phospho- and total levels of Akt and Erk1/2 in

MSCs after stimulation with CMMSC and CMOMSC with or without the addition of PDGFR tyrosine

kinase inhibitor. (B) and (C) Quantitative measurement of the protein band density of phospho-Akt and

phospho-Erk1/2. Compared to CMMSC, CMOMSC induced a substantial phosphorylation of Akt. The pre-

treatment of PDGFR tyrosine kinase inhibitor attenuated the Akt phosphorylation induced by CMOMSC.

No significant difference was noticed in the phosphorylation status of Erk1/2 in MSCs after different

treatments (*p<0.05). CMMSC, conditioned medium derived from undifferentiated mesenchymal

stromal cells; CMOMSC, conditioned medium derived from osteogenically differentiated mesenchymal

stromal cells; PDGFRi, PDGFR tyrosine kinase inhibitor.

Page 117: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

97 | P a g e

Figure 27. Activation of the PI3K/Akt signaling is required for CMOMSC

induced

MSCs migration. (A) Western blotting evaluation of phospho- and total levels of Akt in CMOMSC-

stimulated MSCs at different time points with or without the pre-treatment of 10 µM LY294002. (B)

Quantitative measurement of the protein band density of phospho-Akt. CMOMSC induced the activity of

Akt within 30 min at highest level with gradual decrease till 60 min after stimulation. LY294002, a

specific PI3K/Akt inhibitor, suppressed the CMOMSC

-induced phosphorylation of Akt; (C) In vitro

migration assay showed that CMOMSC-induced MSC migration was significantly interfered by the

addition of LY294002 (*p<0.05). CMOMSC, conditioned medium derived from osteogenically

differentiated mesenchymal stromal cells.

Page 118: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

98 | P a g e

Activation of the PI3K/Akt signaling pathway downstream effectors

The phosphorylation status of downstream effectors of the PI3K/Akt signaling

pathway was evaluated by Western blotting analysis. As shown in Figure 28 A and B,

CMOMSC

significantly increased the phosphorylation of mTOR, p70S6K

, and PAK1 in

MSCs, compared to serum-free medium and CMMSC

. However, the phosphorylation

status of mTOR, p70S6K

, and PAK1 was dramatically suppressed in response to the

pre-treatment of LY294002.

Figure 28 The phosphorylation status of downstream effectors of the PI3K/Akt

signaling pathway. (A) Western blotting evaluation of phospho- and total levels of mTOR, p70S6K

and PAK1 in MSCs in response to various stimulations. (B) Quantitative measurement of the protein

band density of phospho-mTOR, phospho-p70S6K, and phospho-PAK1. CMOMSC induced significantly

increased phosphorylation of mTOR, p70S6K, and PAK1 in MSCs, compared to SF and CMMSC.

LY294002 suppressed the CMOMSC-induced phosphorylation of mTOR, p70S6K, and PAK1 (*p<0.05).

SF, serum-free medium; CMMSC, conditioned medium derived from undifferentiated mesenchymal

stromal cells; CMOMSC, conditioned medium derived from osteogenically differentiated mesenchymal

stromal cells.

Page 119: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

99 | P a g e

DISCUSSION

Targeted migration of endogenous cells to the required site is an essential aspect for

potential therapeutic success. However, the means by which donor cells trigger the

necessary signals required for the recruitment of host MSCs to the defect sites have

remained a puzzling gap in knowledge. For the last decade, considerable effort has

been directed toward identifying the signaling pathways that contribute to host cell

migration and the regeneration of damaged tissues. Our previous studies (REPORT

ONE and REPORT TWO) have shown that donor OMSCs attract host MSCs by

releasing VEGF among a variety of growth factors. Only little is known, however,

about how VEGF secreted by OMSCs signal through the downstream pathways that

instruct the recruitment of MSCs.

As a member from the VEGF/PDGF family, VEGF is the potent mitogen for

endothelial cells (ECs). VEGF not only regulates angiogenesis/vasculogenesis and

vascular permeability, but also induces and enhances ECs proliferation and migration

[374]. Numerous studies have suggested that the chemoattractant activity of VEGF

could be mediated through VEGFR-1, VEGFR-2 and VEGFR-3 [375-378]. However,

a number of studies have reported that limited VEGFR expression was found in

MSCs, indicating VEGF may signal through other receptors in MSCs [200, 372, 379].

In fact, previous reports have also demonstrated that VEGF and PDGF members are

closely related, and VEGFRs and PDGFRs exhibit structural similarities [223, 371].

The PDGF ligands exert their biological activities via two distinct but structurally

related receptors, PDGF receptor-α (PDGFR-α) and PDGFR-β. Particularly strong

expression of PDGFR-α has been noticed in subtypes of mesenchymal progenitors in

lung, skin and intestine, while PDGFR-β is reported to express in the mesenchyme,

especially in vascular SMCs (vSMCs) and pericytes [222]. The present study revealed

that although MSCs only expressed low levels of VEGFR-1, VEGFR-2 and VEGFR-

3, they did express high levels of PDGFR-α and PDGFR-β, which were activated by

the stimulation of VEGF and CM derived from OMSCs. Furthermore, blocking the

biological activity of PDGFR-α and PDGFR-β in MSCs by PDGFR tyrosine kinase

inhibitor prevented MSCs migration induced by CMOMSC

. However, the function of

VEGFRs had no impact on MSC directional migration. Our results suggested that

Page 120: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

100 | P a g e

functional cell surface PDGFR-α and PDGFR-β are both crucial determinants in

mediating MSC migration induced by OMSCs.

Having demonstrated that VEGF promotes mobilization of MSCs towards OMSCs

via PDGFRs, a series of experiments were performed to identify the downstream

signaling pathways participating in this process. The PI3K/Akt signal transduction has

been shown to be a crucial element in multiple cellular function, including cell

survival, proliferation, migration and invasion induced by VEGF among other

cytokines or growth factors [380]. This knowledge prompted us to investigate the

PI3K/Akt signaling in the context of what influence OMSCs have on the motility of

MSCs. The protein kinase Akt, a multifunctional regulator of cell survival, is the key

downstream effector of PI3K. One of the major functions of Akt is to phosphorylate

and activate mTOR, which is a protein kinase of the PI3K/Akt signaling pathway with

a central role in the regulation of cell proliferation, survival and mobility [381].

Subsequently, activated mTOR regulates p70S6K

and PAK1 phosphorylation. The Akt-

mTOR-p70S6K

-PAK1 cascade has been considered not only a central regulatory

pathway of the protein translation involved in regulating cell proliferation, growth,

differentiation and survival, but also a crucial step leading to actin cytoskeleton

reorganization. In the present study, detailed pathway analyses revealed that the

interaction of OMSCs with MSCs led to a rapidly increased PI3K/Akt

phosphorylation via the activation of PDGFRs. We also observed that the downstream

effectors of the PI3K/Akt signaling pathway, mTOR, p70S6K

and PAK1 were

significantly activated when MSCs were stimulated with CMOMSC

. However, the

pharmacological suppression of the PI3K/Akt activity resulted in a complete

attenuation of CMOMSC

-induced cell morphology change. These results suggested that

the PI3K/Akt kinase was activated and served as an essential downstream signaling

molecule in OMSCs-induced MSCs migration.

CONCLUSION

In summary, we demonstrated that MSCs possess potent migratory capacity after

stimulated by OMSCs, which can be applied in clinical setting to maximize the

regeneration effects of the cell-based therapeutic approaches. Our results also

indicated that the PI3K/Akt signaling pathway plays an important role in the

Page 121: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

101 | P a g e

recruitment of MSCs. More intensive studies are required to identify the responsive

host cells and to further explore the cellular and molecular mechanisms that govern

the directional migration of the host cells, thereby allowing for optimisation of the

therapeutic potential of pre-osteogenic differentiation of donor cells to be employed

for bone regeneration.

Page 122: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

102 | P a g e

Page 123: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

103 | P a g e

CHAPTER 5 CONCLUSIONS AND PERSPECTIVES

Page 124: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

104 | P a g e

5.1 General discussion

Bone is a dynamic, mineralized tissue that continuously undergoes modeling and

remodeling, and possesses the intrinsic capacity for regeneration in response to injury

[382]. However, large bone defects resulting from trauma, tumor, and infection

cannot heal properly without intervention, and thus the treatment for these skeletal

deficits represents a major challenge in orthopaedic medicine. Although autologous

bone grafting is widely used in the clinic and has been the gold standard due to its

osteogenic, osteoinductive, and osteoconductive capacity [383], it is often associated

with limited tissue availability, chronic pain, and considerable morbidity at the donor

site [384, 385]. Allogenic or xenogenic bone grafting can be alternative options, but

these strategies have certain drawbacks that include inferior osteoactivity compared to

autologous bone grafting, donor incompatibility, and an increased risk of pathogen

transmission [383, 386].

In view of the limitations of conventional bone graft strategies, tissue engineering

recently has emerged as a promising approach for bone repair and regeneration. The

fundamental concept of bone tissue engineering is to combine progenitor cells, such

as MSCs or osteogenically differentiated/mature cells (for osteogenesis) seeded onto

biocompatible scaffolds and ideally in three-dimensional structures (for

osteoconduction and vascular ingrowth), with appropriate growth factors (for

osteoinduction) to trigger and utilize the endogenous biological capacity to generate

functional bone structures.

5.1.1 Scaffold materials for bone tissue engineering

Tissue engineering scaffolds with a well-defined architecture are designed to

influence the physical, chemical and biological environment to facilitate cell

attachment, proliferation and differentiation. Growth factors and other biomolecules

can be incorporated into the scaffold to guide the regulation of cellular function

during tissue regeneration [387]. The application of the tissue engineering scaffolds

provide the temporary support structure for tissue forming cells to synthesize desired

tissue and to allow nutrient diffusion. Scaffolds for bone tissue engineering are

Page 125: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

105 | P a g e

commonly constructed from biodegradable polymeric materials, either synthetic or

natural [388]. However, for the regeneration of load-bearing bones, the use of

biodegradable polymer scaffolds is challenging because of their low mechanical

strength. Attempts have been made to reinforce the biodegradable polymers with

biocompatible inorganic materials, such as calcium phosphate-based bioceramics and

bioactive glass.

5.1.1.1 Ceramics and bioactive glasses

Ceramics have long been recognized as potent osteoconductive materials for bone

tissue engineering. Calcium phosphate-based ceramics, such as hydroxyapatite (HA)

and β-tricalcium phosphate (β-TCP) are the inorganic materials which attract most

attention for bone repair application. When compared to β-TCP, HA shows minimal

biodegradation, which blocks the formation of new bones and remodeling, and results

in poor local stability or permanent stress concentration [389]. β-TCP is relatively

balanced between bone formation and scaffold absorption. It is also a good

biodegradable ceramic material which can supply a large quantity of calcium ion and

sulfate ion for bone regeneration [389]. However, the mechanical strength of β-TCP is

relatively weaker than that of HA [390, 391]. Furthermore, single β-TCP lacks of

osteoinductivity and osteogenicity, which restricts its application in bone tissue

engineering [389, 391]. In order to optimize the application of both HA and β-TCP,

some researchers have manipulated different ratio of a more stable phase (HA) and a

more soluble one (β-TCP), allowing the formation of biphasic calcium phosphate

(BCP) with a preferable degradation rate and mechanical properties [392].

Ever since the report of the bone-bonding properties of 45S5 Bioglass® more than 40

years ago [393], bioactive glasses (BG) based on silicate [394], phosphate [395] or

borate [396] represent another essential group of inorganic, bioactive scaffold

materials used for bone tissue engineering. The mechanism of the bonding of BG to

bone has been attributed to the formation of an HA-like layer on the glass surface in

contact with the body fluid [397]. It is believed that the initial formation of HA-like

Page 126: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

106 | P a g e

layer involves the adsorption of growth factors, followed by attachment, proliferation

and differentiation of osteoprogenitor cells [390]. 45S5 Bioglass® has been the gold

standard for the application of BG in bone tissue engineering, however, as a scaffold

material, it has several limitations. The difficulty of processing 45S5 Bioglass®

into

porous 3D scaffolds results in low mechanical strength [398]. Furthermore, the low

degradation rate of 45S5 Bioglass®

makes it difficult to match the rate of new bone

tissue formation [397]. The conversion of the scaffold to an HA-like layer is often

incomplete, thus a portion of unconverted glass remains in the scaffold, raising

uncertainty about its effect on the local microenvironment in vivo.

5.1.1.2 Synthetic degradable polymers

Synthetic degradable polymers possess various advantages such as predictable batch

to batch uniformity, being free from concerns of immunogenicity, and having a

reliable source of raw material [388]. Polyglycolide (PGA), polylactide (PLA) and

poly(lactide-co-glycolide) (PLGA) belong to the saturated aliphatic polymeric

materials, which are one of the ancient and most frequently used groups of materials

in the field of bone tissue engineering.

PGA is a rigid, thermoplastic material with a highly crystallinity (45-55%), and

therefore it exhibits high tensile modules with extremely low solubility in organic

solvents [399]. Due to its excellent fiber forming ability, PGA was initially

investigated as one of the best candidates for absorbable sutures. The first

biodegradable synthetic suture, Dexon™, which was approved by the US Food and

Drug Administration in the 1960s, was based on PGA. Sutures made from PGA lose

about 50% of their strength after two weeks, 100% after four weeks and are

completely absorbed within 4-6 months [400]. Non-woven PGA fabrics have been

extensively used as scaffold materials for tissue regeneration due to their excellent

degradability and good initial mechanical properties. However, the accumulation of

the degradation product, glycolic acid, may worsen the pH environment surrounding

Page 127: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

107 | P a g e

the cells and cause damage to the tissues, which may limit the biomedical application

of PGA.

PLA is synthesized by the dimerization of lactic acid. Lactic acid is a chiral molecule

with two optical isomers, the naturally occurring L (Levorotatory) isomer, and the D

(Dextrorotatory) isomer. Therefore, the polymers are referred to as PLLA and PDLA,

respectively. There is also a racemic mixture of D,L-PLA named PDLLA [401]. PLLA

is more commonly used, because the biodegradation product of PLLA is naturally

available in the human body. PLA has linear structure, and one pendent methyl group

which makes it more amorphous and hydrophobic than PGA [388]. However, being

more hydrophobic, the degradation rate of PLA is relatively low. It has been reported

that high molecular weight PLLA can take more than five years to be completely

absorbed, whereas about one year is needed for PDLLA [402]. Being a low strength

polymer with faster degradation rate compared to PLLA, PDLLA is the preferred

candidate for developing drug delivery vehicles and as low strength scaffold material

for tissue regeneration.

PLGA is synthesized through co-polymerizing lactide and glycolide, the mechanical

and biodegradation properties of which are based on the ratio of individual monomers

in the composite. Co-polymers of 25-75% L-LA with glycolide are amorphous due to

the disruption of the regularity of the polymer chain. PLGA polymers containing

50:50 ratio of lactide and glycolide show much greater degradation rate than those

containing higher proportion of either of the two monomers [403]. PLGA

demonstrates good cell adhesion and proliferation, making it a favorable candidate for

tissue engineering applications, especially in blood vessel, bone and cartilage

regeneration [404-406]. A major concern about the application of the synthetic

polymers is the chemicals (additives, traces of catalysts and inhibitors) and monomers

(lactic acid and glycolic acid) released from the degradation of polymer, resulting in

local and systemic host reactions that may cause clinical complications [407]. One of

the by-products, lactic acid, is reported to potentially create an adverse cellular

response at the implant site by reducing the local pH, leading to the release of

Page 128: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

108 | P a g e

prostaglandin E2 (PGE2), a bone resorbing and inflammatory mediator, by human

synovial fibroblasts and murine macrophages [408]. The toxic residues resulting from

the chemical cross-linking also make these synthetic polymers less desirable for

implantable devices [409, 410].

5.1.1.3 Natural degradable polymers

Natural polymers can be considered as the first biodegradable scaffold biomaterials

used clinically. Most of the naturally occurring polymers undergo enzymatic

degradation, and their degradation rate varies significantly with the site of

implantation, depending on the availability and concentration of the enzymes. The

most commonly investigated natural scaffold materials include alginate, collagen,

hyaluronic acid, fibrin, silk and chitosan. Much of the interest in these natural

polymers stems from their superior biocompatibility, relative abundance, the ability to

present receptor-binding ligands to cells, the susceptibility to cell-triggered proteolytic

degradation, and the possibility to mimic the innate microenvironment for bone tissue

regeneration.

Collagen is the most abundant protein in mammals, constituting up to one-third of

total body protein [411]. The individual polypeptide chains of collagen contain 20

different amino acids and the precise composition varies among different tissues [412].

Variations in the amino acid sequence give rise to the different types of collagen,

among which type I collagen is the most predominant type found in the extracellular

matrix (ECM), especially in tissues such as tendon, periosteum and bone [413].

Collagen has been among the most widely used biomaterials for bone tissue

engineering due to its abundance, biocompatibility, high porosity, low antigenicity

and absorbability in the body [414, 415].

The main disadvantage of collagen is its poor mechanical strength, especially

compared with native bone. To enhance its mechanical property, collagen is cross-

Page 129: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

109 | P a g e

linked by a variety of chemical agents or physical treatments. However, the

commonly used chemical agents such as glutaraldehyde and polyepoxy are cytotoxic

at certain concentrations, thus their use has been limited due to the residual cross-

linking compounds remaining in the collagen scaffold [416]. On the other hand,

overexposure to physical treatments, such as dehydrothermal treatment, ultraviolet

irradiation, gamma irradiation and microwave irradiation may lead to collagen

degradation [415]. Another major concern about the application of allogenic or

xenogenic collagen is its potential antigenicity and immunogenicity [417]. However,

reports have shown that the adverse immunological responses only occur in rare cases

and generally settle within a few months, especially in the application of type I

collagen [417, 418]. Premature absorption and other adverse effects of collagen

implantation have been rare and the combined use with immunosuppressant has been

shown to be effective [419].

5.1.2 Growth factors delivery

With improved understanding of fracture healing and bone regeneration at the

molecular level [135], recent research has demonstrated that the incorporation or

release of growth factors, such as basic fibroblast growth factor (bFGF), bone

morphogenetic proteins (BMPs), insulin-like growth factor (IGF), platelet-derived

growth factor (PDGF), transforming growth factor β (TGF-β), and vascular

endothelial growth factor (VEGF) can improve the biological performance of

scaffolds as cell carriers. Of these molecules, BMPs are the most extensively studied,

as they are potent osteoinductive factors. More than 30 BMPs have been identified so

far. Of the known BMPs, BMP-2, -4, and -7 are each individually able to induce new

bone formation at ectopic and orthotopic sites [78, 420-424]. However, the clinical

use of BMPs has been limited because of the expensive and difficult purification

process. Furthermore, the proteins isolated from allogenic or xenogenic sources may

carry potential health risks. The application of recombinant human BMP (rhBMP) has

been reported in a number of animal models as well as a few human clinical trials

[425-427]. The therapeutic potential of rhBMPs-2, -4, and -7 has been proven in

selected fracture repair and bone regeneration process. However, the safety issues

Page 130: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

110 | P a g e

risen from the supraphysiological concentrations of growth factors needed to obtain

the desired osteoinductive effects, the high cost of treatment, and more importantly,

the possibility of local inflammation and edema [428], ectopic bone formation [429]

and carcinogenicity [430] may have reduced the clinical research enthusiasm.

Unlike bone graft transplants where there is a pre-existing vascular supply, synthetic

bone constructs are devoid of vasculature. Researchers have been trying to address

this issue of whether it is a pre-vascularised scaffold developed in vitro or the release

of angiogenic factors from scaffold that promote angiogenesis in situ that will

enhance optimal bone regeneration. VEGF has been one of the most used growth

factors in the application of bone tissue engineering, because it is the key regulator in

a cascade of molecular and cellular events that ultimately lead to the development of

the vascular system [365]. Even though supplying VEGF alone was not sufficient to

initiate the cascade of bone regeneration in the critical-sized calvarial defects, it has

been reported that the combination of angiogenic and osteogenic factors can stimulate

bone healing [431]. Dual release of VEGF and BMP-2 from biodegradable scaffolds

seeded with BMSCs has been demonstrated to be effective for bone repair and

regeneration in a murine femur bone defect model [432]. PLGA scaffolds containing

plasmids encoding DNA for BMP-4 and VEGF have also shown greater bone forming

ability when implanted subcutaneously [433]. Hence, the combined delivery system

of growth factors at different rate kinetics locally from biodegradable scaffolds may

be a promising approach to enhance the reparative mechanism of bone defects.

Although these findings are interesting, the possible application of VEGF in bone

tissue engineering is limited by the short biological half-life, the need for repeated

examinations for optimum dosage, and the expensive cost for the provision of a

sustained concentration at the site of bone regeneration.

5.1.3 Cell sources

Recent advances in stem cell biology and regenerative medicine enable the

development of cell-based therapy. Cell-based bone tissue engineering approaches

entail the seeding of a scaffold with progenitor cells or mature cells that will be

Page 131: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

111 | P a g e

delivered to and retained within the lesion site. It has been reported that stem cell

delivery is likely to produce more reliable and effective results in the management of

bone defect compared to the traditional treatments [434, 435]. MSCs can be easily

isolated from readily accessible bone marrow among other tissues. Additionally, in

vitro expansion potential enables the generation of a sufficient number of bone

marrow-derived MSCs for transplantation [436]. Immunosuppressive and anti-

inflammatory functions [437, 438], homing ability to injured sites [439, 440] and

capability to modify the microenvironment by paracrine factors [441] enable MSCs to

become a promising candidate for cell-based bone tissue engineering.

It has been noted that stem cells alone are not sufficient for a long lasting regenerative

effect. The acceleration of endogenous regenerative mechanism that involves host cell

recruitment, a biological process also known as cell homing, has been considered as a

highly useful and practical approach for bone tissue regeneration [151, 310].

Although there are reports claiming that the MSCs from systemic sources which

express CXCR4 at their surface actively participate in bone regeneration [442, 443], it

is generally accepted that the skeletal progenitors within the local environment play a

more predominant role [444]. These skeletal progenitors may come from the bone

marrow within the injured bone, from the surrounding periosteum, and from soft

tissues in close proximity with the bone. However, all these tissues are closely linked,

which makes it difficult to distinguish their participation during bone repair and

regeneration.

There is a significant body of evidence that demonstrates the transplanted cells can

either differentiate into bone forming cells or attract other cell types to build up bone

tissue and simultaneously resorb cell carrier or bone substitute material [445-447].

Thus, the development of donor cell delivery approaches which can establish key

interactions with host cells in ways that unlock the endogenous power for robust

regeneration becomes increasingly important. The systemic delivery of donor cells

does not require invasive surgical intervention, and the donor cells could potentially

be injected at multiple time points. Furthermore, systemic delivery may be more

Page 132: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

112 | P a g e

beneficial to certain bone disorders affecting the whole body [448, 449]. However,

when delivered systemically, the donor cells contribute modestly to bone forming

cells [444]. Therefore, systemic delivery may not be ideal to direct exogenous cells

toward osteogenesis in a bone defect site. To augment the therapeutic effects of donor

cells, a number of strategies have been developed to deliver donor cells locally and

support bone regeneration. It has been reported that the volume of mineralized callus

is related with the number and concentration of fibroblast colony-forming units in the

bone marrow graft [450], indicating that the number of donor cells may play a part in

the osteogenic stimulation. Osteogenic differentiation of donor cells prior to

implantation may favor superior bone regeneration [19, 451, 452], but it decreases the

expansion capacity. Many efforts are also focused on the design of scaffolds to create

a biocompatible environment and provide proper vascularization. Scaffolds loaded

with angiogenic growth factors have been employed initially [453, 454]. Currently,

the implantation of composites of MSCs mixed with endothelial progenitor cells

(EPCs) is considered as a promising alternative [455].

5.1.4 Animal models for bone repair investigations

It is generally accepted that an ideal animal model for bone repair and regeneration

should mimic clinical conditions of bone injury, be able to utilize fixation, allow

mechanical load, create a permissive microenvironment for diffusion of nutrients and

growth factors, and permit angiogenesis [456]. The model of ectopic bone formation

is commonly employed because it is the least invasive and more suitable for

preliminary screening of donor cells, scaffolds and growth factors [457]. Nearly any

mammal can be chosen, from mouse and rat to rabbit, dog, and pig among numerous

others. To date, the rodent model is the most popular and widely used due to the low

cost, lax skin and availability of immunodeficient animals. A wide variety of locations

have been used for ectopic bone formation model, including subcutaneous,

intramuscular, and kidney capsule transplantation [458]. Subcutaneous implantation is

the simplest of all experimental models of ectopic bone formation. However, the

subcutaneous model is largely devoid of appropriate mechanical stimulation, which

may lead to eventual degradation of the newly formed bone. The physical

Page 133: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

113 | P a g e

identification of the implant can be challenging, especially when the newly formed

bone shares similar color with the surrounding dermal tissues. The bone forming

capacity is actually inferior in subcutaneous models compared to other ectopic models

due to reduced vascularization and blood flow. The lax skin may also allow

potentially significant migration of the implants, making it difficult for sample

collection.

For non-weight bearing testing, the bone defect can be created within the calvaria, rib

or mandibles, which have comparatively low mechanical forces. In this model, most

scaffold materials can be designed to fit the size of the defect and do not require

further fixation. For weight bearing testing, the defect can be made in long bones,

such as femur or tibia, using an osteotomy approach or a traumatic approach [456].

Osteotomy can surgically remove the required length of bone from a predetermined

site, producing a consistent defect in all subjects. However, this does not reflect the

actual clinical conditions following traumatic injury which produce a jagged cut edge

and traumatize both the bone and surrounding soft tissue. The disadvantage of the

traumatic approach is the potential for larger variation in defect size among subjects.

In a weight bearing site, the bone defect is reconstructed with the test material alone

or in combination with cells and growth factors and fixed using external or internal

fixation. Since the animal will start to move after recovery, the biomechanical

properties of the selected scaffold are crucial for optimum bone regeneration in a

weight bearing bone defect. Scaffolds need to be strong enough to allow weight-

bearing during the initial stage of the regenerative process but also biodegradable at

the right time to allow bone remodeling [457]. The contribution of host and donor

cells to bone formation in these complex animal models still remains to be elucidated.

5.2 Project summary

At the time when this project was first conceived, very few attempts had been made to

understand the relationship between host cells and donor cells, although an apparent

correlation between these two counterparts is present at a clinical scale [130-132].

The project sought to advance the knowledge and understanding of the interactions

Page 134: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

114 | P a g e

between donor cells and host cells during osteogenesis, the involvement of host cells

in the bone healing process, and the role of pre-programmed donor cells in the

recruitment of host cells. The ultimate goal is to unveil the potential molecular

mechanisms involved during this process at a cellular level. This project was designed

with three separate but closely–linked segments, which in the end led to three

independent reports summarized as follows.

Firstly, in REPORT ONE, we highlighted the concept of cell-based therapy and the

discrepancies that exist in the literature with respect to the interaction and

involvement of host cells and donor cells during osteogenesis. Having demonstrated

the enhanced migration tendency of MSCs towards OMSCs in the in vitro migration

assay, we then further investigated the role of donor cells during osteogenesis in a

small animal model. Using type-I collagen as a cell-carrier, donor cells were

implanted subcutaneously or into skull defects. Pre-manipulation of the donor cells

was found to significantly enhance mineralization and bone formation. The most

interesting finding was that in both ectopic and orthotopic sites, the implantation of

OMSCs could attract host cells and initiate osteogenesis. These findings provide some

insight into the paracrine effect of the secreted factors from OMSCs on the host cell

recruitment in vivo.

Using a combined in situ hybridisation and immunohistochemical staining technique,

our histological images revealed that the donor OMSCs strongly expressed VEGF,

giving a clue that VEGF secreted by donor OMSCs may play a role in the host cell

recruitment. Therefore, in REPORT TWO we particularly focused on the potential

effect of VEGF. Our data showed that VEGF secretion increased dramatically in

MSCs after osteogenic differentiation in vitro. The secretion of VEGF promoted the

motility of MSCs with elongated actin filament and enhanced the expression of

CXCL12/CXCR4, which is believed to be the most important axis that contributes to

MSCs homing to areas of damaged tissue. In vivo findings revealed that blocking

VEGF with neutralizing antibody led to a remarkable decrease of MSC recruitment

and new bone formation. Thus, the results from this study for the first time showed

Page 135: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

115 | P a g e

that VEGF secreted by transplanted OMSCs altered the cell morphology and

CXCL12/CXCR4 expression pattern of MSCs, triggering the downstream signaling

pathway, which may explain the host cell recruitment phenomenon observed in our

previous study.

In REPORT THREE, we first demonstrated that MSCs express limited functional

VEGFRs, which is in agreement with other reports [200, 372, 379]. As VEGF and

PDGF are closely related members from the VEGF/PDGF super family, VEGFRs and

PDGFRs are also structurally related, so we speculated that VEGF may exert its

paracrine effect through the activation of PDGFRs. Therefore, we furthered our study

and the results showed that the function of VEGFRs had no impact on MSC

directional migration, instead, the functional cell surface PDGFRs were crucial

determinants in mediating MSC migration induced by OMSCs. In order to unveil the

root causes of the enhanced host cell recruitment, our study probed cell signaling

pathways that are highly related to cell polarization and migration, such as PI3K/Akt

and Erk1/2. Our data showed that OMSCs induced a swift activation of PI3K/Akt in

MSCs, leading to the phosphorylation of downstream effectors, such as mTOR,

p70S6K

, and PAK1. This segment of study showed that the VEGF secreted from

OMSCs promoted MSCs directional migration via the activation of PDGFRs, which

then led to the activation of the PI3K/Akt signaling pathway.

5.3 Future direction

In this thesis, we mainly focused on the paracrine effect of VEGF secreted from

OMSCs, however, there are other cytokines derived from OMSCs which may also

play a significant role in host cell recruitment. Therefore, further studies are justified

to identify such soluble factors, probably by using proteomic mapping of the

conditioned medium derived from OMSCs. Also, during the last decade, growing

evidence has indicated that preferential activation of PI3K/Akt at the side of the cell

facing the chemoattractant gradient is critical for establishing a new leading edge, cell

polarization, and directional movement in a wide range of motile cell types [240, 459,

460]. Thus the clear recognition that the PI3K/Akt signaling pathway underlines the

Page 136: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

116 | P a g e

recruitment of host cells towards bone defect sites makes the clarification of

downstream targets of central importance to our understanding of bone defect healing.

More advanced animal models, such as knock-ins encoding catalytically inactive Akt

mutants, will likely provide a more accurate picture of the role of the PI3K/Akt

signaling pathway in bone tissue regeneration driven by the recruited endogenous

cells. It will also be important to further define the role of crosstalk between distinct

downstream branches of the PI3K/Akt signaling pathway, as well as the networking

with other signaling pathways, in the ultimate outcome of optimal bone remodelling

and regeneration.

Taken together, the observations from this work suggest that host cells and donor cells

interact closely in the bone healing process after cell transplantation. We have

reported for the first time that the implanted OMSCs play an integral role in recruiting

host cells towards the implantation sites, and more importantly, we have demonstrated

the factors secreted from donor cells (e.g. VEGF) can induce cell morphology

changes and enhance expression of CXCL12/CXCR4. We also reported that VEGF

secreted by OMSCs can signal through PDGFRs in MSCs. The signal is then

amplified by the activation of the PI3K/Akt kinase, ultimately leading to endogenous

cell recruitment and robust bone regeneration. This highlights the novelty of the

obtained results and therefore provides the scientific rationale to define new

therapeutic approaches for bone defect healing.

Page 137: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

117 | P a g e

REFERENCES 1. Krasny K, Kaminski A, Krasny M, Zadurska M, Piekarczyk P, Fiedor P. Clinical use of

allogeneic bone granulates to reconstruct maxillary and mandibular alveolar processes. Transplant Proc 2011. 43(8): p. 3142-4.

2. Kim YK, Yun PY, Lee HJ, Ahn JY, Kim SG. Ridge preservation of the molar extraction socket using collagen sponge and xenogeneic bone grafts. Implant Dent 2011. 20(4): p. 267-72.

3. Kolk A, Handschel J, Drescher W, et al. Current trends and future perspectives of bone substitute materials - From space holders to innovative biomaterials. J Craniomaxillofac Surg 2012. 40(8): p. 706-18.

4. Zhao L, Zhao J, Wang S, Wang J, Liu J. Comparative study between tissue-engineered periosteum and structural allograft in rabbit critical-sized radial defect model. J Biomed Mater Res B Appl Biomater 2011. 97(1): p. 1-9.

5. Yang HL, Zhu XS, Chen L, et al. Bone healing response to a synthetic calcium sulfate/beta-tricalcium phosphate graft material in a sheep vertebral body defect model. J Biomed Mater Res B Appl Biomater 2012. 100B(7): p. 1911-21.

6. Brownfield LA, Weltman RL. Ridge preservation with or without an osteoinductive allograft: a clinical, radiographic, micro-computed tomography, and histologic study evaluating dimensional changes and new bone formation of the alveolar ridge. J Periodontol 2012. 83(5): p. 581-9.

7. Herford AS, Dean JS. Complications in bone grafting. Oral Maxillofac Surg Clin North Am 2011. 23(3): p. 433-42.

8. Neman J, Hambrecht A, Cadry C, Jandial R. Stem cell-mediated osteogenesis: therapeutic potential for bone tissue engineering. Biologics 2012. 6: p. 47-57.

9. Zimmermann JA, Knothe Tate ML. Structure-function relationships in the stem cell's mechanical world A: seeding protocols as a means to control shape and fate of live stem cells. Mol Cell Biomech 2011. 8(4): p. 275-96.

10. Kim K, Dean D, Lu A, Mikos AG, Fisher JP. Early osteogenic signal expression of rat bone marrow stromal cells is influenced by both hydroxyapatite nanoparticle content and initial cell seeding density in biodegradable nanocomposite scaffolds. Acta Biomater 2011. 7(3): p. 1249-64.

11. Grayson WL, Bhumiratana S, Cannizzaro C, et al. Effects of initial seeding density and fluid perfusion rate on formation of tissue-engineered bone. Tissue Eng Part A 2008. 14(11): p. 1809-20.

12. Willems WF, Larsen M, Friedrich PF, Shogren KL, Bishop AT. Induction of angiogenesis and osteogenesis in surgically revascularized frozen bone allografts by sustained delivery of FGF-2 and VEGF. J Orthop Res 2012. 30(10): p. 1556-62.

13. Behr B, Tang C, Germann G, Longaker MT, Quarto N. Locally applied vascular endothelial growth factor A increases the osteogenic healing capacity of human adipose-derived stem cells by promoting osteogenic and endothelial differentiation. Stem Cells 2011. 29(2): p. 286-96.

14. Lee JH, Um S, Jang JH, Seo BM. Effects of VEGF and FGF-2 on proliferation and differentiation of human periodontal ligament stem cells. Cell Tissue Res 2012. 348(3): p. 475-84.

15. D' Alimonte I, Nargi E, Mastrangelo F, et al. Vascular endothelial growth factor enhances in vitro proliferation and osteogenic differentiation of human dental pulp stem cells. J Biol Regul Homeost Agents 2011. 25(1): p. 57-69.

Page 138: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

118 | P a g e

16. Tsai CC, Chen YJ, Yew TL, et al. Hypoxia inhibits senescence and maintains mesenchymal stem cell properties through down-regulation of E2A-p21 by HIF-TWIST. Blood 2011. 117(2): p. 459-69.

17. Hung SP, Ho JH, Shih YR, Lo T, Lee OK. Hypoxia promotes proliferation and osteogenic differentiation potentials of human mesenchymal stem cells. J Orthop Res 2012. 30(2): p. 260-6.

18. Hirao M, Hashimoto J, Yamasaki N, et al. Oxygen tension is an important mediator of the transformation of osteoblasts to osteocytes. J Bone Miner Metab 2007. 25(5): p. 266-76.

19. Ye X, Yin X, Yang D, Tan J, Liu G. Ectopic bone regeneration by human bone marrow mononucleated cells, undifferentiated and osteogenically differentiated bone marrow mesenchymal stem cells in beta-tricalcium phosphate scaffolds. Tissue Eng Part C Methods 2012. 18(7): p. 545-56.

20. Han D, Sun X, Zhang X, Tang T, Dai K. Ectopic osteogenesis by ex vivo gene therapy using beta tricalcium phosphate as a carrier. Connect Tissue Res 2008. 49(5): p. 343-50.

21. Kang SW, Lee JS, Park MS, Park JH, Kim BS. Enhancement of in vivo bone regeneration efficacy of human mesenchymal stem cells. J Microbiol Biotechnol 2008. 18(5): p. 975-82.

22. Lee JY, Musgrave D, Pelinkovic D, et al. Effect of bone morphogenetic protein-2-expressing muscle-derived cells on healing of critical-sized bone defects in mice. J Bone Joint Surg Am 2001. 83-A(7): p. 1032-9.

23. Xiao Y, Qian H, Young WG, Bartold PM. Tissue engineering for bone regeneration using differentiated alveolar bone cells in collagen scaffolds. Tissue Eng 2003. 9(6): p. 1167-77.

24. Greco SJ, Rameshwar P. Microenvironmental considerations in the application of human mesenchymal stem cells in regenerative therapies. Biologics 2008. 2(4): p. 699-705.

25. Panetta NJ, Gupta DM, Quarto N, Longaker MT. Mesenchymal cells for skeletal tissue engineering. Panminerva Med 2009. 51(1): p. 25-41.

26. Fox JM, Chamberlain G, Ashton BA, Middleton J. Recent advances into the understanding of mesenchymal stem cell trafficking. Br J Haematol 2007. 137(6): p. 491-502.

27. Langer R, Vacanti JP. Tissue engineering. Science 1993. 260(5110): p. 920-6. 28. Zhou H, Guo M, Bian C, et al. Efficacy of bone marrow-derived mesenchymal stem

cells in the treatment of sclerodermatous chronic graft-versus-host disease: clinical report. Biol Blood Marrow Transplant 2010. 16(3): p. 403-12.

29. Lunde K, Solheim S, Aakhus S, et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med 2006. 355(12): p. 1199-209.

30. Yerebakan C, Kaminski A, Westphal B, et al. Impact of preoperative left ventricular function and time from infarction on the long-term benefits after intramyocardial CD133(+) bone marrow stem cell transplant. J Thorac Cardiovasc Surg 2011. 142(6): p. 1530-9

31. Kharaziha P, Hellstrom PM, Noorinayer B, et al. Improvement of liver function in liver cirrhosis patients after autologous mesenchymal stem cell injection: a phase I-II clinical trial. Eur J Gastroenterol Hepatol 2009. 21(10): p. 1199-205.

32. Frolov AA, Bryukhovetskiy AS. Effects of hematopoietic autologous stem cell transplantation to the chronically injured human spinal cord evaluated by motor and

Page 139: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

119 | P a g e

somatosensory evoked potentials methods. Cell Transplant 2012. 21 (Suppl 1): p. S49-55.

33. Cashman N, Tan LY, Krieger C, et al. Pilot study of granulocyte colony stimulating factor (G-CSF)-mobilized peripheral blood stem cells in amyotrophic lateral sclerosis (ALS). Muscle Nerve 2008. 37(5): p. 620-5.

34. Clarke AW, Alyas F, Morris T, Robertson CJ, Bell J, Connell DA. Skin-derived tenocyte-like cells for the treatment of patellar tendinopathy. Am J Sports Med 2011. 39(3): p. 614-23.

35. Wakitani S, Okabe T, Horibe S, et al. Safety of autologous bone marrow-derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months. J Tissue Eng Regen Med 2011. 5(2): p. 146-50.

36. Nagata M, Hoshina H, Li M, et al. A clinical study of alveolar bone tissue engineering with cultured autogenous periosteal cells: coordinated activation of bone formation and resorption. Bone 2012. 50(5): p. 1123-9.

37. Paul J, Sagstetter A, Kriner M, Imhoff AB, Spang J, Hinterwimmer S. Donor-site morbidity after osteochondral autologous transplantation for lesions of the talus. J Bone Joint Surg Am 2009. 91(7): p. 1683-8.

38. Dimitriou R, Jones E, McGonagle D, Giannoudis PV. Bone regeneration: current concepts and future directions. BMC Med 2011. 9: p. 66-76.

39. Viateau V, Guillemin G, Bousson V, et al. Long-bone critical-size defects treated with tissue-engineered grafts: a study on sheep. J Orthop Res 2007. 25(6): p. 741-9.

40. Liu G, Zhao L, Zhang W, Cui L, Liu W, Cao Y. Repair of goat tibial defects with bone marrow stromal cells and beta-tricalcium phosphate. J Mater Sci Mater Med 2008. 19(6): p. 2367-76.

41. Cao L, Liu X, Liu S, et al. Experimental repair of segmental bone defects in rabbits by angiopoietin-1 gene transfected MSCs seeded on porous beta-TCP scaffolds. J Biomed Mater Res B Appl Biomater 2012. 100(5): p. 1229-36.

42. Sauerbier S, Stricker A, Kuschnierz J, et al. In vivo comparison of hard tissue regeneration with human mesenchymal stem cells processed with either the FICOLL method or the BMAC method. Tissue Eng Part C Methods 2010. 16(2): p. 215-23.

43. Yamada Y, Nakamura S, Ito K, et al. Injectable tissue-engineered bone using autogenous bone marrow-derived stromal cells for maxillary sinus augmentation: clinical application report from a 2-6-year follow-up. Tissue Eng Part A 2008. 14(10): p. 1699-707.

44. Loh YH, Wu Q, Chew JL, et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet 2006. 38(4): p. 431-40.

45. Cole MF, Johnstone SE, Newman JJ, Kagey MH, Young RA. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells. Genes Dev 2008. 22(6): p. 746-55.

46. Jiang J, Chan YS, Loh YH, et al. A core Klf circuitry regulates self-renewal of embryonic stem cells. Nat Cell Biol 2008. 10(3): p. 353-60.

47. Kim J, Chu J, Shen X, Wang J, Orkin SH. An extended transcriptional network for pluripotency of embryonic stem cells. Cell 2008. 132(6): p. 1049-61.

48. Logeart-Avramoglou D, Anagnostou F, Bizios R, Petite H. Engineering bone: challenges and obstacles. J Cell Mol Med 2005. 9(1): p. 72-84.

49. Faour O, Dimitriou R, Cousins CA, Giannoudis PV. The use of bone graft substitutes in large cancellous voids: any specific needs? Injury 2011. 42 Suppl 2: p. S87-90.

Page 140: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

120 | P a g e

50. Graham SM, Leonidou A, Aslam-Pervez N, et al. Biological therapy of bone defects: the immunology of bone allo-transplantation. Expert Opin Biol Ther 2010. 10(6): p. 885-901.

51. Ward WG, Gautreaux MD, Lippert DC, 2nd, Boles C. HLA sensitization and allograft bone graft incorporation. Clin Orthop Relat Res 2008. 466(8): p. 1837-48.

52. Markel DC, Guthrie ST, Wu B, Song Z, Wooley PH. Characterization of the inflammatory response to four commercial bone graft substitutes using a murine biocompatibility model. J Inflamm Res 2012. 5: p. 13-8.

53. Robinson D, Alk D, Sandbank J, Farber R, Halperin N. Inflammatory reactions associated with a calcium sulfate bone substitute. Ann Transplant 1999. 4(3-4): p. 91-7.

54. Castro S, II, Zambuzzi WF, de Oliveira Castro L, Granjeiro JM. Periosteal-derived cells for bone bioengineering: a promising candidate. Clin Oral Implants Res 2012. 23(10): p. 1238-42.

55. Wang F, Yu M, Yan X, et al. Gingiva-derived mesenchymal stem cell-mediated therapeutic approach for bone tissue regeneration. Stem Cells Dev 2011. 20(12): p. 2093-102.

56. Yang Y, Hallgrimsson B, Putnins EE. Craniofacial defect regeneration using engineered bone marrow mesenchymal stromal cells. J Biomed Mater Res A 2011. 99(1): p. 74-85.

57. Davatchi F, Abdollahi BS, Mohyeddin M, Shahram F, Nikbin B. Mesenchymal stem cell therapy for knee osteoarthritis. Preliminary report of four patients. Int J Rheum Dis 2011. 14(2): p. 211-5.

58. Marcacci M, Kon E, Moukhachev V, et al. Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study. Tissue Eng 2007. 13(5): p. 947-55.

59. Chen FM, Wu LA, Zhang M, Zhang R, Sun HH. Homing of endogenous stem/progenitor cells for in situ tissue regeneration: Promises, strategies, and translational perspectives. Biomaterials 2011. 32(12): p. 3189-209.

60. Daley GQ, Scadden DT. Prospects for stem cell-based therapy. Cell 2008. 132(4): p. 544-8.

61. Teo AK, Vallier L. Emerging use of stem cells in regenerative medicine. Biochem J 2010. 428(1): p. 11-23.

62. Jungebluth P, Alici E, Baiguera S, et al. Tracheobronchial transplantation with a stem-cell-seeded bioartificial nanocomposite: a proof-of-concept study. Lancet 2011. 378(9808): p. 1997-2004.

63. Ye KY, Black LD, 3rd. Strategies for tissue engineering cardiac constructs to affect functional repair following myocardial infarction. J Cardiovasc Transl Res 2011. 4(5): p. 575-91.

64. Leach JK, Mooney DJ. Bone engineering by controlled delivery of osteoinductive molecules and cells. Expert Opin Biol Ther 2004. 4(7): p. 1015-27.

65. Serigano K, Sakai D, Hiyama A, Tamura F, Tanaka M, Mochida J. Effect of cell number on mesenchymal stem cell transplantation in a canine disc degeneration model. J Orthop Res 2010. 28(10): p. 1267-75.

66. Wu J, Li J, Zhang N, Zhang C. Stem cell-based therapies in ischemic heart diseases: a focus on aspects of microcirculation and inflammation. Basic Res Cardiol 2011. 106(3): p. 317-24.

67. Wuertz K, Godburn K, Iatridis JC. MSC response to pH levels found in degenerating intervertebral discs. Biochem Biophys Res Commun 2009. 379(4): p. 824-9.

Page 141: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

121 | P a g e

68. Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant 2011. 20(1): p. 5-14.

69. Harkness L, Mahmood A, Ditzel N, Abdallah BM, Nygaard JV, Kassem M. Selective isolation and differentiation of a stromal population of human embryonic stem cells with osteogenic potential. Bone 2011. 48(2): p. 231-41.

70. Cohen CB. Ethical and policy issues surrounding the donation of cryopreserved and fresh embryos for human embryonic stem cell research. Stem Cell Rev 2009. 5(2): p. 116-22.

71. Slater BJ, Kwan MD, Gupta DM, Panetta NJ, Longaker MT. Mesenchymal cells for skeletal tissue engineering. Expert Opin Biol Ther 2008. 8(7): p. 885-93.

72. Manini I, Gulino L, Gava B, et al. Multi-potent progenitors in freshly isolated and cultured human mesenchymal stem cells: a comparison between adipose and dermal tissue. Cell Tissue Res 2011. 344(1): p. 85-95.

73. Bajpai VK, Mistriotis P, Andreadis ST. Clonal multipotency and effect of long-term in vitro expansion on differentiation potential of human hair follicle derived mesenchymal stem cells. Stem Cell Res 2012. 8(1): p. 74-84.

74. Kisiel AH, McDuffee LA, Masaoud E, Bailey TR, Esparza Gonzalez BP, Nino-Fong R. Isolation, characterization, and in vitro proliferation of canine mesenchymal stem cells derived from bone marrow, adipose tissue, muscle, and periosteum. Am J Vet Res 2012. 73(8): p. 1305-17.

75. Chen D, Shen H, Shao J, et al. Superior mineralization and neovascularization capacity of adult human metaphyseal periosteum-derived cells for skeletal tissue engineering applications. Int J Mol Med 2011. 27(5): p. 707-13.

76. Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells 2007. 25(6): p. 1384-92.

77. Liu G, Li Y, Sun J, et al. In vitro and in vivo evaluation of osteogenesis of human umbilical cord blood-derived mesenchymal stem cells on partially demineralized bone matrix. Tissue Eng Part A 2010. 16(3): p. 971-82.

78. Zhang Y, Fan W, Nothdurft L, et al. In vitro and in vivo evaluation of adenovirus combined silk fibroin scaffolds for bone morphogenetic protein-7 gene delivery. Tissue Eng Part C Methods 2011. 17(8): p. 789-97.

79. Zhao J, Shen G, Liu C, et al. Enhanced healing of rat calvarial defects with sulfated chitosan-coated calcium-deficient hydroxyapatite/bone morphogenetic protein 2 scaffolds. Tissue Eng Part A 2012. 18(1-2): p. 185-97.

80. Meinel L, Hofmann S, Betz O, et al. Osteogenesis by human mesenchymal stem cells cultured on silk biomaterials: comparison of adenovirus mediated gene transfer and protein delivery of BMP-2. Biomaterials 2006. 27(28): p. 4993-5002.

81. Wang X, Cui F, Madhu V, Dighe AS, Balian G, Cui Q. Combined VEGF and LMP-1 delivery enhances osteoprogenitor cell differentiation and ectopic bone formation. Growth Factors 2011. 29(1): p. 36-48.

82. Cui F, Wang X, Liu X, Dighe AS, Balian G, Cui Q. VEGF and BMP-6 enhance bone formation mediated by cloned mouse osteoprogenitor cells. Growth Factors 2010. 28(5): p. 306-17.

83. Bellantuono I, Aldahmash A, Kassem M. Aging of marrow stromal (skeletal) stem cells and their contribution to age-related bone loss. Biochim Biophys Acta 2009. 1792(4): p. 364-70.

84. Asumda FZ, Chase PB. Age-related changes in rat bone-marrow mesenchymal stem cell plasticity. BMC Cell Biol 2011. 12: p. 44.

Page 142: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

122 | P a g e

85. Benisch P, Schilling T, Klein-Hitpass L, et al. The transcriptional profile of mesenchymal stem cell populations in primary osteoporosis is distinct and shows overexpression of osteogenic inhibitors. PLoS One 2012. 7(9): p. e45142.

86. Mafi P, Hindocha S, Mafi R, Griffin M, Khan WS. Adult mesenchymal stem cells and cell surface characterization - a systematic review of the literature. Open Orthop J 2011. 5(Suppl 2): p. 253-60.

87. Rojewski MT, Weber BM, Schrezenmeier H. Phenotypic Characterization of Mesenchymal Stem Cells from Various Tissues. Transfus Med Hemother 2008. 35(3): p. 168-84.

88. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 2007. 25(11): p. 2739-49.

89. Barry F, Boynton R, Murphy M, Haynesworth S, Zaia J. The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells. Biochem Biophys Res Commun 2001. 289(2): p. 519-24.

90. Barry FP, Boynton RE, Haynesworth S, Murphy JM, Zaia J. The monoclonal antibody SH-2, raised against human mesenchymal stem cells, recognizes an epitope on endoglin (CD105). Biochem Biophys Res Commun 1999. 265(1): p. 134-9.

91. Nadri S, Soleimani M, Hosseni RH, Massumi M, Atashi A, Izadpanah R. An efficient method for isolation of murine bone marrow mesenchymal stem cells. Int J Dev Biol 2007. 51(8): p. 723-9.

92. Modder UI, Roforth MM, Nicks KM, et al. Characterization of mesenchymal progenitor cells isolated from human bone marrow by negative selection. Bone 2012. 50(3): p. 804-10.

93. Majumdar MK, Keane-Moore M, Buyaner D, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci 2003. 10(2): p. 228-41.

94. Kundu AK, Putnam AJ. Vitronectin and collagen I differentially regulate osteogenesis in mesenchymal stem cells. Biochem Biophys Res Commun 2006. 347(1): p. 347-57.

95. Prowse AB, Chong F, Gray PP, Munro TP. Stem cell integrins: implications for ex-vivo culture and cellular therapies. Stem Cell Res 2011. 6(1): p. 1-12.

96. Pimton P, Sarkar S, Sheth N, et al. Fibronectin-mediated upregulation of alpha5beta1 integrin and cell adhesion during differentiation of mouse embryonic stem cells. Cell Adh Migr 2011. 5(1): p. 73-82.

97. Lemieux JM, Horowitz MC, Kacena MA. Involvement of integrins alpha(3)beta(1) and alpha(5)beta(1) and glycoprotein IIb in megakaryocyte-induced osteoblast proliferation. J Cell Biochem 2010. 109(5): p. 927-32.

98. Mruthyunjaya S, Manchanda R, Godbole R, Pujari R, Shiras A, Shastry P. Laminin-1 induces neurite outgrowth in human mesenchymal stem cells in serum/differentiation factors-free conditions through activation of FAK-MEK/ERK signaling pathways. Biochem Biophys Res Commun 2010. 391(1): p. 43-8.

99. Walenda T, Bork S, Horn P, et al. Co-culture with mesenchymal stromal cells increases proliferation and maintenance of haematopoietic progenitor cells. J Cell Mol Med 2010. 14(1-2): p. 337-50.

100. Bianco P, Sacchetti B, Riminucci M. Osteoprogenitors and the hematopoietic microenvironment. Best Pract Res Clin Haematol 2011. 24(1): p. 37-47.

101. Saleh FA, Whyte M, Genever PG. Effects of endothelial cells on human mesenchymal stem cell activity in a three-dimensional in vitro model. Eur Cell Mater 2011. 22: p. 242-57; discussion 57.

Page 143: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

123 | P a g e

102. Becerra J, Santos-Ruiz L, Andrades JA, Mari-Beffa M. The stem cell niche should be a key issue for cell therapy in regenerative medicine. Stem Cell Rev 2011. 7(2): p. 248-55.

103. Schraufstatter IU, Discipio RG, Khaldoyanidi S. Mesenchymal stem cells and their microenvironment. Front Biosci 2011. 16: p. 2271-88.

104. Abdallah BM, Kassem M. Human mesenchymal stem cells: from basic biology to clinical applications. Gene Ther 2008. 15(2): p. 109-16.

105. Lanfer B, Seib FP, Freudenberg U, et al. The growth and differentiation of mesenchymal stem and progenitor cells cultured on aligned collagen matrices. Biomaterials 2009. 30(30): p. 5950-8.

106. Augello A, De Bari C. The regulation of differentiation in mesenchymal stem cells. Hum Gene Ther 2010. 21(10): p. 1226-38.

107. Kollmer M, Keskar V, Hauk TG, Collins JM, Russell B, Gemeinhart RA. Stem cell-derived extracellular matrix enables survival and multilineage differentiation within superporous hydrogels. Biomacromolecules 2012. 13(4): p. 963-73.

108. Kagami H, Agata H, Tojo A. Bone marrow stromal cells (bone marrow-derived multipotent mesenchymal stromal cells) for bone tissue engineering: basic science to clinical translation. Int J Biochem Cell Biol 2011. 43(3): p. 286-9.

109. Mezey E. The therapeutic potential of bone marrow-derived stromal cells. J Cell Biochem 2011. 112(10): p. 2683-7.

110. Min WK, Bae JS, Park BC, et al. Proliferation and osteoblastic differentiation of bone marrow stem cells: comparison of vertebral body and iliac crest. Eur Spine J 2010. 19(10): p. 1753-60.

111. Xue Y, Danmark S, Xing Z, et al. Growth and differentiation of bone marrow stromal cells on biodegradable polymer scaffolds: an in vitro study. J Biomed Mater Res A 2010. 95(4): p. 1244-51.

112. Aghaloo TL, Chaichanasakul T, Bezouglaia O, et al. Osteogenic potential of mandibular vs. long-bone marrow stromal cells. J Dent Res 2010. 89(11): p. 1293-8.

113. Zou D, Zhang Z, Ye D, et al. Repair of critical-sized rat calvarial defects using genetically engineered bone marrow-derived mesenchymal stem cells overexpressing hypoxia-inducible factor-1alpha. Stem Cells 2011. 29(9): p. 1380-90.

114. Guo J, Meng Z, Chen G, et al. Restoration of critical size defects in the rabbit mandible using porous nano-hydroxyapatite-polyamide scaffolds. Tissue Eng Part A 2012. 18(11-12): p. 1239-52.

115. Quarto R, Mastrogiacomo M, Cancedda R, et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med 2001. 344(5): p. 385-6.

116. Cancedda R, Mastrogiacomo M, Bianchi G, Derubeis A, Muraglia A, Quarto R. Bone marrow stromal cells and their use in regenerating bone. Novartis Found Symp 2003. 249: p. 133-43.

117. Wall ST, Yeh CC, Tu RY, Mann MJ, Healy KE. Biomimetic matrices for myocardial stabilization and stem cell transplantation. J Biomed Mater Res A 2010. 95(4): p. 1055-66.

118. Yang Y, Rossi FM, Putnins EE. Periodontal regeneration using engineered bone marrow mesenchymal stromal cells. Biomaterials 2010. 31(33): p. 8574-82.

119. Moosmann S, Hutter J, Moser C, Krombach F, Huss R. Milieu-adopted in vitro and in vivo differentiation of mesenchymal tissues derived from different adult human CD34-negative progenitor cell clones. Cells Tissues Organs 2005. 179(3): p. 91-101.

120. Harris CT, Cooper LF. Comparison of bone graft matrices for human mesenchymal stem cell-directed osteogenesis. J Biomed Mater Res A 2004. 68(4): p. 747-55.

Page 144: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

124 | P a g e

121. Zhang C. Transcriptional regulation of bone formation by the osteoblast-specific transcription factor Osx. J Orthop Surg Res 2010. 5: p. 37.

122. Ducy P, Karsenty G. Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene. Mol Cell Biol 1995. 15(4): p. 1858-69.

123. Komori T. Regulation of osteoblast differentiation by Runx2. Adv Exp Med Biol 2010. 658: p. 43-9.

124. Komori T. Regulation of osteoblast differentiation by transcription factors. J Cell Biochem 2006. 99(5): p. 1233-9.

125. Marie PJ. Transcription factors controlling osteoblastogenesis. Arch Biochem Biophys 2008. 473(2): p. 98-105.

126. Matsuo K. Cross-talk among bone cells. Curr Opin Nephrol Hypertens 2009. 18(4): p. 292-7.

127. Reichert JC, Quent VM, Noth U, Hutmacher DW. Ovine cortical osteoblasts outperform bone marrow cells in an ectopic bone assay. J Tissue Eng Regen Med 2011. 5(10): p. 831-44.

128. Fodor WL. Tissue engineering and cell based therapies, from the bench to the clinic: the potential to replace, repair and regenerate. Reprod Biol Endocrinol 2003. 1: p. 102.

129. Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther 2009. 17(6): p. 939-46.

130. Horwitz EM, Prockop DJ, Fitzpatrick LA, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 1999. 5(3): p. 309-13.

131. Horwitz EM, Gordon PL, Koo WK, et al. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc Natl Acad Sci U S A 2002. 99(13): p. 8932-7.

132. Otsuru S, Gordon PL, Shimono K, et al. Transplanted bone marrow mononuclear cells and MSCs impart clinical benefit to children with osteogenesis imperfecta through different mechanisms. Blood 2012. 120(9): p. 1933-41.

133. Sato T, Iso Y, Uyama T, et al. Coronary vein infusion of multipotent stromal cells from bone marrow preserves cardiac function in swine ischemic cardiomyopathy via enhanced neovascularization. Lab Invest 2011. 91(4): p. 553-64.

134. Iso Y, Spees JL, Serrano C, et al. Multipotent human stromal cells improve cardiac function after myocardial infarction in mice without long-term engraftment. Biochem Biophys Res Commun 2007. 354(3): p. 700-6.

135. Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury 2005. 36(12): p. 1392-404.

136. Zhang ZY, Teoh SH, Hui JH, Fisk NM, Choolani M, Chan JK. The potential of human fetal mesenchymal stem cells for off-the-shelf bone tissue engineering application. Biomaterials 2012. 33(9): p. 2656-72.

137. Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury. Am J Physiol Renal Physiol 2007. 292(5): p. F1626-35.

138. Burdon TJ, Paul A, Noiseux N, Prakash S, Shum-Tim D. Bone marrow stem cell derived paracrine factors for regenerative medicine: current perspectives and therapeutic potential. Bone Marrow Res 2011. 2011: p. 207326.

139. Ratajczak MZ, Kucia M, Jadczyk T, et al. Pivotal role of paracrine effects in stem cell therapies in regenerative medicine: can we translate stem cell-secreted paracrine

Page 145: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

125 | P a g e

factors and microvesicles into better therapeutic strategies? Leukemia 2012. 26(6): p. 1166-73.

140. Gharaibeh B, Lavasani M, Cummins JH, Huard J. Terminal differentiation is not a major determinant for the success of stem cell therapy - cross-talk between muscle-derived stem cells and host cells. Stem Cell Res Ther 2011. 2(4): p. 31.

141. Tortelli F, Tasso R, Loiacono F, Cancedda R. The development of tissue-engineered bone of different origin through endochondral and intramembranous ossification following the implantation of mesenchymal stem cells and osteoblasts in a murine model. Biomaterials 2010. 31(2): p. 242-9.

142. Wu Y, Wang J, Scott PG, Tredget EE. Bone marrow-derived stem cells in wound healing: a review. Wound Repair Regen 2007. 15(Suppl 1): p. S18-26.

143. Kinnaird T, Stabile E, Burnett MS, et al. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res 2004. 94(5): p. 678-85.

144. Moldovan NI. Role of monocytes and macrophages in adult angiogenesis: a light at the tunnel's end. J Hematother Stem Cell Res 2002. 11(2): p. 179-94.

145. Constantin G, Marconi S, Rossi B, et al. Adipose-derived mesenchymal stem cells ameliorate chronic experimental autoimmune encephalomyelitis. Stem Cells 2009. 27(10): p. 2624-35.

146. Diaz-Flores L, Jr., Gutierrez R, Madrid JF, Varela H, Valladares F, Diaz-Flores L. Adult stem cells and repair through granulation tissue. Front Biosci 2009. 14: p. 1433-70.

147. Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, and forces combine and control stem cells. Science 2009. 324(5935): p. 1673-7.

148. Votteler M, Kluger PJ, Walles H, Schenke-Layland K. Stem cell microenvironments--unveiling the secret of how stem cell fate is defined. Macromol Biosci 2010. 10(11): p. 1302-15.

149. Laird DJ, von Andrian UH, Wagers AJ. Stem cell trafficking in tissue development, growth, and disease. Cell 2008. 132(4): p. 612-30.

150. Lee SJ, Van Dyke M, Atala A, Yoo JJ. Host cell mobilization for in situ tissue regeneration. Rejuvenation Res 2008. 11(4): p. 747-56.

151. Lee CH, Cook JL, Mendelson A, Moioli EK, Yao H, Mao JJ. Regeneration of the articular surface of the rabbit synovial joint by cell homing: a proof of concept study. Lancet 2010. 376(9739): p. 440-8.

152. Ko IK, Ju YM, Chen T, Atala A, Yoo JJ, Lee SJ. Combined systemic and local delivery of stem cell inducing/recruiting factors for in situ tissue regeneration. FASEB J 2012. 26(1): p. 158-68.

153. Tasso R, Fais F, Reverberi D, Tortelli F, Cancedda R. The recruitment of two consecutive and different waves of host stem/progenitor cells during the development of tissue-engineered bone in a murine model. Biomaterials 2010. 31(8): p. 2121-9.

154. Shirley D, Marsh D, Jordan G, McQuaid S, Li G. Systemic recruitment of osteoblastic cells in fracture healing. J Orthop Res 2005. 23(5): p. 1013-21.

155. Chodniewicz D, Klemke RL. Guiding cell migration through directed extension and stabilization of pseudopodia. Exp Cell Res 2004. 301(1): p. 31-7.

156. Cramer LP. Forming the cell rear first: breaking cell symmetry to trigger directed cell migration. Nat Cell Biol 2010. 12(7): p. 628-32.

157. Ridley AJ, Schwartz MA, Burridge K, et al. Cell migration: integrating signals from front to back. Science 2003. 302(5651): p. 1704-9.

158. Lauffenburger DA, Horwitz AF. Cell migration: a physically integrated molecular process. Cell 1996. 84(3): p. 359-69.

Page 146: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

126 | P a g e

159. Laing KJ, Secombes CJ. Chemokines. Dev Comp Immunol 2004. 28(5): p. 443-60. 160. Broxmeyer HE, Kim CH, Cooper SH, Hangoc G, Hromas R, Pelus LM. Effects of CC,

CXC, C, and CX3C chemokines on proliferation of myeloid progenitor cells, and insights into SDF-1-induced chemotaxis of progenitors. Ann N Y Acad Sci 1999. 872: p. 142-62; discussion 63.

161. Han ZC, Lu M, Li J, et al. Platelet factor 4 and other CXC chemokines support the survival of normal hematopoietic cells and reduce the chemosensitivity of cells to cytotoxic agents. Blood 1997. 89(7): p. 2328-35.

162. Lataillade JJ, Clay D, Dupuy C, et al. Chemokine SDF-1 enhances circulating CD34(+) cell proliferation in synergy with cytokines: possible role in progenitor survival. Blood 2000. 95(3): p. 756-68.

163. Nomiyama H, Mera A, Ohneda O, Miura R, Suda T, Yoshie O. Organization of the chemokine genes in the human and mouse major clusters of CC and CXC chemokines: diversification between the two species. Genes Immun 2001. 2(2): p. 110-3.

164. Lau TT, Wang DA. Stromal cell-derived factor-1 (SDF-1): homing factor for engineered regenerative medicine. Expert Opin Biol Ther 2011. 11(2): p. 189-97.

165. Tashiro K, Tada H, Heilker R, Shirozu M, Nakano T, Honjo T. Signal sequence trap: a cloning strategy for secreted proteins and type I membrane proteins. Science 1993. 261(5121): p. 600-3.

166. Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 2006. 25(6): p. 977-88.

167. Maijenburg MW, van der Schoot CE, Voermans C. Mesenchymal stromal cell migration: possibilities to improve cellular therapy. Stem Cells Dev 2012. 21(1): p. 19-29.

168. Mendt M, Cardier JE. Stromal-derived factor-1 and its receptor, CXCR4, are constitutively expressed by mouse liver sinusoidal endothelial cells: implications for the regulation of hematopoietic cell migration to the liver during extramedullary hematopoiesis. Stem Cells Dev 2012. 21(12): p. 2142-51.

169. Jin SZ, Meng XW, Han MZ, Sun X, Sun LY, Liu BR. Stromal cell derived factor-1 enhances bone marrow mononuclear cell migration in mice with acute liver failure. World J Gastroenterol 2009. 15(21): p. 2657-64.

170. Shinohara K, Greenfield S, Pan H, et al. Stromal cell-derived factor-1 and monocyte chemotactic protein-3 improve recruitment of osteogenic cells into sites of musculoskeletal repair. J Orthop Res 2011. 29(7): p. 1064-9.

171. Theiss HD, Vallaster M, Rischpler C, et al. Dual stem cell therapy after myocardial infarction acts specifically by enhanced homing via the SDF-1/CXCR4 axis. Stem Cell Res 2011. 7(3): p. 244-55.

172. Zaruba MM, Franz WM. Role of the SDF-1-CXCR4 axis in stem cell-based therapies for ischemic cardiomyopathy. Expert Opin Biol Ther 2010. 10(3): p. 321-35.

173. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004. 10(8): p. 858-64.

174. Weidt C, Niggemann B, Kasenda B, Drell TL, Zanker KS, Dittmar T. Stem cell migration: a quintessential stepping stone to successful therapy. Curr Stem Cell Res Ther 2007. 2(1): p. 89-103.

175. Nagasawa T, Hirota S, Tachibana K, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 1996. 382(6592): p. 635-8.

Page 147: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

127 | P a g e

176. Ma Q, Jones D, Borghesani PR, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci U S A 1998. 95(16): p. 9448-53.

177. Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 1998. 393(6685): p. 595-9.

178. Zhu W, Liang G, Huang Z, Doty SB, Boskey AL. Conditional inactivation of the CXCR4 receptor in osteoprecursors reduces postnatal bone formation due to impaired osteoblast development. J Biol Chem 2011. 286(30): p. 26794-805.

179. Dar A, Goichberg P, Shinder V, et al. Chemokine receptor CXCR4-dependent internalization and resecretion of functional chemokine SDF-1 by bone marrow endothelial and stromal cells. Nat Immunol 2005. 6(10): p. 1038-46.

180. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell 2009. 4(3): p. 206-16.

181. Semerad CL, Christopher MJ, Liu F, et al. G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow. Blood 2005. 106(9): p. 3020-7.

182. Sharma M, Afrin F, Satija N, Tripathi RP, Gangenahalli GU. Stromal-derived factor-1/CXCR4 signaling: indispensable role in homing and engraftment of hematopoietic stem cells in bone marrow. Stem Cells Dev 2011. 20(6): p. 933-46.

183. Ghadge SK, Muhlstedt S, Ozcelik C, Bader M. SDF-1alpha as a therapeutic stem cell homing factor in myocardial infarction. Pharmacol Ther 2011. 129(1): p. 97-108.

184. Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr Top Microbiol Immunol 2010. 341: p. 37-58.

185. Shyu WC, Lin SZ, Yang HI, et al. Functional recovery of stroke rats induced by granulocyte colony-stimulating factor-stimulated stem cells. Circulation 2004. 110(13): p. 1847-54.

186. Ryser MF, Ugarte F, Thieme S, Bornhauser M, Roesen-Wolff A, Brenner S. mRNA transfection of CXCR4-GFP fusion--simply generated by PCR-results in efficient migration of primary human mesenchymal stem cells. Tissue Eng Part C Methods 2008. 14(3): p. 179-84.

187. Bhakta S, Hong P, Koc O. The surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivation. Cardiovasc Revasc Med 2006. 7(1): p. 19-24.

188. Cheng Z, Ou L, Zhou X, et al. Targeted migration of mesenchymal stem cells modified with CXCR4 gene to infarcted myocardium improves cardiac performance. Mol Ther 2008. 16(3): p. 571-9.

189. De Clercq E. Recent advances on the use of the CXCR4 antagonist plerixafor (AMD3100, Mozobil) and potential of other CXCR4 antagonists as stem cell mobilizers. Pharmacol Ther 2010. 128(3): p. 509-18.

190. Jujo K, Hamada H, Iwakura A, et al. CXCR4 blockade augments bone marrow progenitor cell recruitment to the neovasculature and reduces mortality after myocardial infarction. Proc Natl Acad Sci U S A 2010. 107(24): p. 11008-13.

191. Ferrara N. VEGF-A: a critical regulator of blood vessel growth. Eur Cytokine Netw 2009. 20(4): p. 158-63.

192. Portal-Nunez S, Lozano D, Esbrit P. Role of angiogenesis on bone formation. Histol Histopathol 2012. 27(5): p. 559-66.

193. Liu B, Li X, Liang G, Liu X. VEGF expression in mesenchymal stem cells promotes bone formation of tissue-engineered bones. Mol Med Report 2011. 4(6): p. 1121-6.

Page 148: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

128 | P a g e

194. Fischerauer E, Heidari N, Neumayer B, Deutsch A, Weinberg AM. The spatial and temporal expression of VEGF and its receptors 1 and 2 in post-traumatic bone bridge formation of the growth plate. J Mol Histol 2011. 42(6): p. 513-22.

195. Stempel J, Fritsch H, Pfaller K, Blumer MJ. Development of articular cartilage and the metaphyseal growth plate: the localization of TRAP cells, VEGF, and endostatin. J Anat 2011. 218(6): p. 608-18.

196. Orlandini M, Spreafico A, Bardelli M, et al. Vascular endothelial growth factor-D activates VEGFR-3 expressed in osteoblasts inducing their differentiation. J Biol Chem 2006. 281(26): p. 17961-7.

197. Clarkin CE, Emery RJ, Pitsillides AA, Wheeler-Jones CP. Evaluation of VEGF-mediated signaling in primary human cells reveals a paracrine action for VEGF in osteoblast-mediated crosstalk to endothelial cells. J Cell Physiol 2008. 214(2): p. 537-44.

198. Street J, Bao M, deGuzman L, et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Pro Natl Acad Sci U S A 2002. 99(15): p. 9656-61.

199. Deckers MM, Karperien M, van der Bent C, Yamashita T, Papapoulos SE, Lowik CW. Expression of vascular endothelial growth factors and their receptors during osteoblast differentiation. Endocrinology 2000. 141(5): p. 1667-74.

200. Furumatsu T, Shen ZN, Kawai A, et al. Vascular endothelial growth factor principally acts as the main angiogenic factor in the early stage of human osteoblastogenesis. J Biochem 2003. 133(5): p. 633-9.

201. Tang W, Yang F, Li Y, et al. Transcriptional regulation of Vascular Endothelial Growth Factor (VEGF) by osteoblast-specific transcription factor Osterix (Osx) in osteoblasts. J Biol Chem 2012. 287(3): p. 1671-8.

202. Mayr-Wohlfart U, Waltenberger J, Hausser H, et al. Vascular endothelial growth factor stimulates chemotactic migration of primary human osteoblasts. Bone 2002. 30(3): p. 472-7.

203. Fiedler J, Leucht F, Waltenberger J, Dehio C, Brenner RE. VEGF-A and PlGF-1 stimulate chemotactic migration of human mesenchymal progenitor cells. Biochem Biophys Res Commun 2005. 334(2): p. 561-8.

204. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003. 9(6): p. 669-76.

205. Lee S, Chen TT, Barber CL, et al. Autocrine VEGF signaling is required for vascular homeostasis. Cell 2007. 130(4): p. 691-703.

206. Yamazaki Y, Morita T. Molecular and functional diversity of vascular endothelial growth factors. Mol Divers 2006. 10(4): p. 515-27.

207. Holmes DI, Zachary I. The vascular endothelial growth factor (VEGF) family: angiogenic factors in health and disease. Genome Biol 2005. 6(2): p. 209.

208. Neufeld G, Cohen T, Shraga N, Lange T, Kessler O, Herzog Y. The neuropilins: multifunctional semaphorin and VEGF receptors that modulate axon guidance and angiogenesis. Trends Cardiovasc Med 2002. 12(1): p. 13-9.

209. Soker S, Miao HQ, Nomi M, Takashima S, Klagsbrun M. VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J Cell Biochem 2002. 85(2): p. 357-68.

210. Ball SG, Shuttleworth CA, Kielty CM. Vascular endothelial growth factor can signal through platelet-derived growth factor receptors. J Cell Biol 2007. 177(3): p. 489-500.

211. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer 2008. 8(8): p. 579-91.

212. Nazarenko I, Hede SM, He X, et al. PDGF and PDGF receptors in glioma. Ups J Med Sci 2012. 117(2): p. 99-112.

Page 149: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

129 | P a g e

213. Kohler N, Lipton A. Platelets as a source of fibroblast growth-promoting activity. Exp Cell Res 1974. 87(2): p. 297-301.

214. Ross R, Glomset J, Kariya B, Harker L. A platelet-dependent serum factor that stimulates the proliferation of arterial smooth muscle cells in vitro. Proc Natl Acad Sci U S A 1974. 71(4): p. 1207-10.

215. Westermark B, Wasteson A. A platelet factor stimulating human normal glial cells. Exp Cell Res 1976. 98(1): p. 170-4.

216. Tallquist M, Kazlauskas A. PDGF signaling in cells and mice. Cytokine Growth Factor Rev 2004. 15(4): p. 205-13.

217. Li X, Ponten A, Aase K, et al. PDGF-C is a new protease-activated ligand for the PDGF alpha-receptor. Nat Cell Biol 2000. 2(5): p. 302-9.

218. LaRochelle WJ, Jeffers M, McDonald WF, et al. PDGF-D, a new protease-activated growth factor. Nat Cell Biol 2001. 3(5): p. 517-21.

219. Reigstad LJ, Varhaug JE, Lillehaug JR. Structural and functional specificities of PDGF-C and PDGF-D, the novel members of the platelet-derived growth factors family. FEBS J 2005. 272(22): p. 5723-41.

220. Ustach CV, Huang W, Conley-LaComb MK, et al. A novel signaling axis of matriptase/PDGF-D/ss-PDGFR in human prostate cancer. Cancer Res 2010. 70(23): p. 9631-40.

221. Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev 2004. 15(4): p. 197-204.

222. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev 2008. 22(10): p. 1276-312.

223. Ball SG, Shuttleworth CA, Kielty CM. Platelet-derived growth factor receptor-alpha is a key determinant of smooth muscle alpha-actin filaments in bone marrow-derived mesenchymal stem cells. Int J Biochem Cell Biol 2007. 39(2): p. 379-91.

224. Betsholtz C. Biology of platelet-derived growth factors in development. Birth Defects Res C Embryo Today 2003. 69(4): p. 272-85.

225. Ostendorf T, Eitner F, Floege J. The PDGF family in renal fibrosis. Pediatr Nephrol 2012. 27(7): p. 1041-50.

226. Ronnstrand L, Heldin CH. Mechanisms of platelet-derived growth factor-induced chemotaxis. Int J Cancer 2001. 91(6): p. 757-62.

227. Rosenkranz S, DeMali KA, Gelderloos JA, Bazenet C, Kazlauskas A. Identification of the receptor-associated signaling enzymes that are required for platelet-derived growth factor-AA-dependent chemotaxis and DNA synthesis. J Biol Chem 1999. 274(40): p. 28335-43.

228. Nagel M, Tahinci E, Symes K, Winklbauer R. Guidance of mesoderm cell migration in the Xenopus gastrula requires PDGF signaling. Development 2004. 131(11): p. 2727-36.

229. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science 1997. 277(5323): p. 242-5.

230. Hellstrom M, Kalen M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 1999. 126(14): p. 3047-55.

231. French WJ, Creemers EE, Tallquist MD. Platelet-derived growth factor receptors direct vascular development independent of vascular smooth muscle cell function. Mol Cell Biol 2008. 28(18): p. 5646-57.

232. Ferrari G, Cusella-De Angelis G, Coletta M, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 1998. 279(5356): p. 1528-30.

Page 150: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

130 | P a g e

233. Fiedler J, Etzel N, Brenner RE. To go or not to go: Migration of human mesenchymal progenitor cells stimulated by isoforms of PDGF. J Cell Biochem 2004. 93(5): p. 990-8.

234. Tepper OM, Capla JM, Galiano RD, et al. Adult vasculogenesis occurs through in situ recruitment, proliferation, and tubulization of circulating bone marrow-derived cells. Blood 2005. 105(3): p. 1068-77.

235. Veevers-Lowe J, Ball SG, Shuttleworth A, Kielty CM. Mesenchymal stem cell migration is regulated by fibronectin through alpha5beta1-integrin-mediated activation of PDGFR-beta and potentiation of growth factor signals. J Cell Sci 2011. 124(Pt 8): p. 1288-300.

236. Whitman M, Kaplan DR, Schaffhausen B, Cantley L, Roberts TM. Association of phosphatidylinositol kinase activity with polyoma middle-T competent for transformation. Nature 1985. 315(6016): p. 239-42.

237. Jiang BH, Liu LZ. PI3K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer Res 2009. 102: p. 19-65.

238. Sadhu C, Masinovsky B, Dick K, Sowell CG, Staunton DE. Essential role of phosphoinositide 3-kinase delta in neutrophil directional movement. J Immunol 2003. 170(5): p. 2647-54.

239. Schymeinsky J, Then C, Sindrilaru A, et al. Syk-mediated translocation of PI3Kdelta to the leading edge controls lamellipodium formation and migration of leukocytes. PLoS One 2007. 2(11): p. e1132.

240. Yoo SK, Deng Q, Cavnar PJ, Wu YI, Hahn KM, Huttenlocher A. Differential regulation of protrusion and polarity by PI3K during neutrophil motility in live zebrafish. Dev Cell 2010. 18(2): p. 226-36.

241. Li S, Deng L, Gong L, Bian H, Dai Y, Wang Y. Upregulation of CXCR4 favoring neural-like cells migration via AKT activation. Neurosci Res 2010. 67(4): p. 293-9.

242. Yu J, Li M, Qu Z, Yan D, Li D, Ruan Q. SDF-1/CXCR4-mediated migration of transplanted bone marrow stromal cells toward areas of heart myocardial infarction through activation of PI3K/Akt. J Cardiovasc Pharmacol 2010. 55(5): p. 496-505.

243. Vanhaesebroeck B, Leevers SJ, Ahmadi K, et al. Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem 2001. 70: p. 535-602.

244. Thi EP, Reiner NE. Phosphatidylinositol 3-kinases and their roles in phagosome maturation. J Leukoc Biol 2012. 92(3): p. 553-66.

245. Suire S, Coadwell J, Ferguson GJ, Davidson K, Hawkins P, Stephens L. p84, a new Gbetagamma-activated regulatory subunit of the type IB phosphoinositide 3-kinase p110gamma. Curr Biol 2005. 15(6): p. 566-70.

246. Voigt P, Brock C, Nurnberg B, Schaefer M. Assigning functional domains within the p101 regulatory subunit of phosphoinositide 3-kinase gamma. J Biol Chem 2005. 280(6): p. 5121-7.

247. Cain RJ, Ridley AJ. Phosphoinositide 3-kinases in cell migration. Biol Cell 2009. 101(1): p. 13-29.

248. Domin J, Harper L, Aubyn D, et al. The class II phosphoinositide 3-kinase PI3K-C2beta regulates cell migration by a PtdIns3P dependent mechanism. J Cell Physiol 2005. 205(3): p. 452-62.

249. Maffucci T, Cooke FT, Foster FM, Traer CJ, Fry MJ, Falasca M. Class II phosphoinositide 3-kinase defines a novel signaling pathway in cell migration. J Cell Biol 2005. 169(5): p. 789-99.

250. Backer JM. The regulation and function of Class III PI3Ks: novel roles for Vps34. Biochem J 2008. 410(1): p. 1-17.

251. Bunney TD, Katan M. Phosphoinositide signalling in cancer: beyond PI3K and PTEN. Nat Rev Cancer 2010. 10(5): p. 342-52.

Page 151: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

131 | P a g e

252. Yoeli-Lerner M, Toker A. Akt/PKB signaling in cancer: a function in cell motility and invasion. Cell Cycle 2006. 5(6): p. 603-5.

253. Sepe M, Festa L, Tolino F, et al. A regulatory mechanism involving TBP-1/Tat-Binding Protein 1 and Akt/PKB in the control of cell proliferation. PLoS One 2011. 6(10): p. e22800.

254. Miyake H, Maeda K, Asai N, et al. The actin-binding protein Girdin and its Akt-mediated phosphorylation regulate neointima formation after vascular injury. Circ Res 2011. 108(10): p. 1170-9.

255. Diez H, Garrido JJ, Wandosell F. Specific roles of Akt iso forms in apoptosis and axon growth regulation in neurons. PLoS One 2012. 7(4): p. e32715.

256. Kubota H, Noguchi R, Toyoshima Y, et al. Temporal coding of insulin action through multiplexing of the AKT pathway. Mol Cell 2012. 46(6): p. 820-32.

257. Fenouille N, Tichet M, Dufies M, et al. The epithelial-mesenchymal transition (EMT) regulatory factor SLUG (SNAI2) is a downstream target of SPARC and AKT in promoting melanoma cell invasion. PLoS One 2012. 7(7): p. e40378.

258. Dellinger MT, Brekken RA. Phosphorylation of Akt and ERK1/2 is required for VEGF-A/VEGFR2-induced proliferation and migration of lymphatic endothelium. PLoS One 2011. 6(12): p. e28947.

259. Zou W, Chu X, Cai C, et al. AKT-mediated regulation of polarization in differentiated human neutrophil-like HL-60 cells. Inflamm Res 2012. 61(8): p. 853-62.

260. Wang Z, Yang J, Fisher T, Xiao H, Jiang Y, Yang C. Akt activation is responsible for enhanced migratory and invasive behavior of arsenic-transformed human bronchial epithelial cells. Environ Health Perspect 2012. 120(1): p. 92-7.

261. Irie HY, Pearline RV, Grueneberg D, et al. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J Cell Biol 2005. 171(6): p. 1023-34.

262. Yoeli-Lerner M, Yiu GK, Rabinovitz I, Erhardt P, Jauliac S, Toker A. Akt blocks breast cancer cell motility and invasion through the transcription factor NFAT. Mol Cell 2005. 20(4): p. 539-50.

263. Kim EK, Yun SJ, Do KH, et al. Lysophosphatidic acid induces cell migration through the selective activation of Akt1. Exp Mol Med 2008. 40(4): p. 445-52.

264. Kim EK, Yun SJ, Ha JM, et al. Selective activation of Akt1 by mammalian target of rapamycin complex 2 regulates cancer cell migration, invasion, and metastasis. Oncogene 2011. 30(26): p. 2954-63.

265. Zhang B, Gu F, She C, et al. Reduction of Akt2 inhibits migration and invasion of glioma cells. Int J Cancer 2009. 125(3): p. 585-95.

266. Wang J, Wan W, Sun R, et al. Reduction of Akt2 expression inhibits chemotaxis signal transduction in human breast cancer cells. Cell Signal 2008. 20(6): p. 1025-34.

267. Zhou GL, Tucker DF, Bae SS, Bhatheja K, Birnbaum MJ, Field J. Opposing roles for Akt1 and Akt2 in Rac/Pak signaling and cell migration. J Biol Chem 2006. 281(47): p. 36443-53.

268. Virtakoivu R, Pellinen T, Rantala JK, Perala M, Ivaska J. Distinct roles of AKT isoforms in regulating beta1-integrin activity, migration, and invasion in prostate cancer. Mol Biol Cell 2012. 23(17): p. 3357-69.

269. Tomas NM, Masur K, Piecha JC, Niggemann B, Zanker KS. Akt and phospholipase Cgamma are involved in the regulation of growth and migration of MDA-MB-468 breast cancer and SW480 colon cancer cells when cultured with diabetogenic levels of glucose and insulin. BMC Res Notes 2012. 5: p. 214.

Page 152: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

132 | P a g e

270. Rowland AF, Larance M, Hughes WE, James DE. Identification of RhoGAP22 as an Akt-dependent regulator of cell motility in response to insulin. Mol Cell Biol 2011. 31(23): p. 4789-800.

271. Xiong W, Cheng BH, Jia SB, Tang LS. Involvement of the PI3K/Akt signaling pathway in platelet-derived growth factor-induced migration of human lens epithelial cells. Curr Eye Res 2010. 35(5): p. 389-401.

272. Wang CH, Cao GF, Jiang Q, Yao J. TNF-alpha promotes human retinal pigment epithelial (RPE) cell migration by inducing matrix metallopeptidase 9 (MMP-9) expression through activation of Akt/mTORC1 signaling. Biochem Biophys Res Commun 2012. 425(1): p. 33-8.

273. Zheng H, Fu G, Dai T, Huang H. Migration of endothelial progenitor cells mediated by stromal cell-derived factor-1alpha/CXCR4 via PI3K/Akt/eNOS signal transduction pathway. J Cardiovasc Pharmacol 2007. 50(3): p. 274-80.

274. Ryu CH, Park SA, Kim SM, et al. Migration of human umbilical cord blood mesenchymal stem cells mediated by stromal cell-derived factor-1/CXCR4 axis via Akt, ERK, and p38 signal transduction pathways. Biochem Biophys Res Commun 2010. 398(1): p. 105-10.

275. Wu M, Chen Q, Li D, et al. LRRC4 inhibits human glioblastoma cells proliferation, invasion, and proMMP-2 activation by reducing SDF-1 alpha/CXCR4-mediated ERK1/2 and Akt signaling pathways. J Cell Biochem 2008. 103(1): p. 245-55.

276. Delgado-Martin C, Escribano C, Pablos JL, Riol-Blanco L, Rodriguez-Fernandez JL. Chemokine CXCL12 uses CXCR4 and a signaling core formed by bifunctional Akt, extracellular signal-regulated kinase (ERK)1/2, and mammalian target of rapamycin complex 1 (mTORC1) proteins to control chemotaxis and survival simultaneously in mature dendritic cells. J Biol Chem 2011. 286(43): p. 37222-36.

277. Sotsios Y, Whittaker GC, Westwick J, Ward SG. The CXC chemokine stromal cell-derived factor activates a Gi-coupled phosphoinositide 3-kinase in T lymphocytes. J Immunol 1999. 163(11): p. 5954-63.

278. Rosova I, Dao M, Capoccia B, Link D, Nolta JA. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells 2008. 26(8): p. 2173-82.

279. Enomoto A, Murakami H, Asai N, et al. Akt/PKB regulates actin organization and cell motility via Girdin/APE. Dev Cell 2005. 9(3): p. 389-402.

280. Jiang P, Enomoto A, Jijiwa M, et al. An actin-binding protein Girdin regulates the motility of breast cancer cells. Cancer Res 2008. 68(5): p. 1310-8.

281. Ip CK, Cheung AN, Ngan HY, Wong AS. p70 S6 kinase in the control of actin cytoskeleton dynamics and directed migration of ovarian cancer cells. Oncogene 2011. 30(21): p. 2420-32.

282. Jung ID, Lee J, Lee KB, et al. Activation of p21-activated kinase 1 is required for lysophosphatidic acid-induced focal adhesion kinase phosphorylation and cell motility in human melanoma A2058 cells. Eur J Biochem 2004. 271(8): p. 1557-65.

283. Arias-Romero LE, Chernoff J. A tale of two Paks. Biol Cell 2008. 100(2): p. 97-108. 284. Dummler B, Ohshiro K, Kumar R, Field J. Pak protein kinases and their role in cancer.

Cancer Metastasis Rev 2009. 28(1-2): p. 51-63. 285. Delorme V, Machacek M, DerMardirossian C, et al. Cofilin activity downstream of

Pak1 regulates cell protrusion efficiency by organizing lamellipodium and lamella actin networks. Dev Cell 2007. 13(5): p. 646-62.

286. Ching YP, Leong VY, Lee MF, Xu HT, Jin DY, Ng IO. P21-activated protein kinase is overexpressed in hepatocellular carcinoma and enhances cancer metastasis

Page 153: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

133 | P a g e

involving c-Jun NH2-terminal kinase activation and paxillin phosphorylation. Cancer Res 2007. 67(8): p. 3601-8.

287. Brown MC, West KA, Turner CE. Paxillin-dependent paxillin kinase linker and p21-activated kinase localization to focal adhesions involves a multistep activation pathway. Mol Biol Cell 2002. 13(5): p. 1550-65.

288. Rincon M, Davis RJ. Regulation of the immune response by stress-activated protein kinases. Immunol Rev 2009. 228(1): p. 212-24.

289. Brown MD, Sacks DB. Protein scaffolds in MAP kinase signalling. Cell Signal 2009. 21(4): p. 462-9.

290. Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 2004. 68(2): p. 320-44.

291. Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev 2011. 75(1): p. 50-83.

292. Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev 2001. 81(2): p. 807-69.

293. Farooq A, Zhou MM. Structure and regulation of MAPK phosphatases. Cell Signal 2004. 16(7): p. 769-79.

294. Ralph JA, Morand EF. MAPK phosphatases as novel targets for rheumatoid arthritis. Expert Opin Ther Targets 2008. 12(7): p. 795-808.

295. Pearson G, Robinson F, Beers Gibson T, et al. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 2001. 22(2): p. 153-83.

296. Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta 2010. 1802(4): p. 396-405.

297. Huang C, Jacobson K, Schaller MD. MAP kinases and cell migration. J Cell Sci 2004. 117(Pt 20): p. 4619-28.

298. Meng XN, Jin Y, Yu Y, et al. Characterisation of fibronectin-mediated FAK signalling pathways in lung cancer cell migration and invasion. Br J Cancer 2009. 101(2): p. 327-34.

299. Klemke RL, Cai S, Giannini AL, Gallagher PJ, de Lanerolle P, Cheresh DA. Regulation of cell motility by mitogen-activated protein kinase. J Cell Biol 1997. 137(2): p. 481-92.

300. Ma L, Lan F, Zheng Z, et al. Epidermal growth factor (EGF) and interleukin (IL)-1beta synergistically promote ERK1/2-mediated invasive breast ductal cancer cell migration and invasion. Mol Cancer 2012. 11(1): p. 79.

301. Shi C, Lu J, Wu W, et al. Endothelial cell-specific molecule 2 (ECSM2) localizes to cell-cell junctions and modulates bFGF-directed cell migration via the ERK-FAK pathway. PLoS One 2011. 6(6): p. e21482.

302. Kagami S, Urushihara M, Kitamura A, et al. PDGF-BB enhances alpha1beta1 integrin-mediated activation of the ERK/AP-1 pathway involved in collagen matrix remodeling by rat mesangial cells. J Cell Physiol 2004. 198(3): p. 470-8.

303. Zhao R, Liu XQ, Wu XP, et al. Vascular endothelial growth factor (VEGF) enhances gastric carcinoma invasiveness via integrin alpha(v)beta6. Cancer Lett 2010. 287(2): p. 150-6.

304. Degryse B, Orlando S, Resnati M, Rabbani SA, Blasi F. Urokinase/urokinase receptor and vitronectin/alpha(v)beta(3) integrin induce chemotaxis and cytoskeleton reorganization through different signaling pathways. Oncogene 2001. 20(16): p. 2032-43.

Page 154: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

134 | P a g e

305. Jo M, Thomas KS, Somlyo AV, Somlyo AP, Gonias SL. Cooperativity between the Ras-ERK and Rho-Rho kinase pathways in urokinase-type plasminogen activator-stimulated cell migration. J Biol Chem 2002. 277(14): p. 12479-85.

306. Liu Y, Shepherd EG, Nelin LD. MAPK phosphatases--regulating the immune response. Nat Rev Immunol 2007. 7(3): p. 202-12.

307. Jethva R, Otsuru S, Dominici M, Horwitz EM. Cell therapy for disorders of bone. Cytotherapy 2009. 11(1): p. 3-17.

308. Lee K, Chan CK, Patil N, Goodman SB. Cell therapy for bone regeneration--bench to bedside. J Biomed Mater Res B Appl Biomater 2009. 89(1): p. 252-63.

309. Shea GK, Tsui AY, Chan YS, Shum DK. Bone marrow-derived Schwann cells achieve fate commitment--a prerequisite for remyelination therapy. Exp Neurol 2010. 224(2): p. 448-58.

310. Zhao J, Zhang N, Prestwich GD, Wen X. Recruitment of endogenous stem cells for tissue repair. Macromol Biosci 2008. 8(9): p. 836-42.

311. Xiao Y, Haase H, Young WG, Bartold PM. Development and transplantation of a mineralized matrix formed by osteoblasts in vitro for bone regeneration. Cell Transplant 2004. 13(1): p. 15-25.

312. Lin CY, Chang YH, Lin KJ, et al. The healing of critical-sized femoral segmental bone defects in rabbits using baculovirus-engineered mesenchymal stem cells. Biomaterials 2010. 31(12): p. 3222-30.

313. Watanabe N, Takai S, Morita N, Kawata M, Hirasawa Y. A method of tracking donor cells after simulated autologous transplantation: a study using synovial cells of transgenic rats. Cell Tissue Res 1999. 298(3): p. 519-25.

314. Lacerda SA, Lanzoni JF, Bombonato-Prado KF, Campos AA, Prata CA, Brentegani LG. Osteogenic potential of autogenous bone associated with bone marrow osteoblastic cells in bony defects: a histomorphometric study. Implant Dent 2009. 18(6): p. 521-9.

315. Shao Z, Liu B, Peng Q, et al. Transplantation of osteoblast-like cells to the distracted callus in the rabbit mandible. Plast Reconstr Surg 2007. 119(2): p. 500-7.

316. Li F, Wang X, Niyibizi C. Bone marrow stromal cells contribute to bone formation following infusion into femoral cavities of a mouse model of osteogenesis imperfecta. Bone 2010. 47(3): p. 546-55.

317. Siddappa R, Fernandes H, Liu J, van Blitterswijk C, de Boer J. The response of human mesenchymal stem cells to osteogenic signals and its impact on bone tissue engineering. Curr Stem Cell Res Ther 2007. 2(3): p. 209-20.

318. Mareddy S, Crawford R, Brooke G, Xiao Y. Clonal isolation and characterization of bone marrow stromal cells from patients with osteoarthritis. Tissue Eng 2007. 13(4): p. 819-29.

319. Singh S, Jones BJ, Crawford R, Xiao Y. Characterization of a mesenchymal-like stem cell population from osteophyte tissue. Stem Cells Dev 2008. 17(2): p. 245-54.

320. Singh S, Dhaliwal N, Crawford R, Xiao Y. Cellular senescence and longevity of osteophyte-derived mesenchymal stem cells compared to patient-matched bone marrow stromal cells. J Cell Biochem 2009. 108(4): p. 839-50.

321. Zhang Y, Fan W, Ma Z, et al. The effects of pore architecture in silk fibroin scaffolds on the growth and differentiation of mesenchymal stem cells expressing BMP7. Acta Biomater 2010. 6(8): p. 3021-8.

322. Khan F, Tare RS, Kanczler JM, Oreffo RO, Bradley M. Strategies for cell manipulation and skeletal tissue engineering using high-throughput polymer blend formulation and microarray techniques. Biomaterials 2010. 31(8): p. 2216-28.

323. Friedenstein AJ. Precursor cells of mechanocytes. Int Rev Cytol 1976. 47: p. 327-59.

Page 155: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

135 | P a g e

324. Liu W, Cui L, Cao Y. Bone reconstruction with bone marrow stromal cells. Methods Enzymol 2006. 420: p. 362-80.

325. Docheva D, Popov C, Mutschler W, Schieker M. Human mesenchymal stem cells in contact with their environment: surface characteristics and the integrin system. J Cell Mol Med 2007. 11(1): p. 21-38.

326. Fu L, Tang T, Miao Y, Zhang S, Qu Z, Dai K. Stimulation of osteogenic differentiation and inhibition of adipogenic differentiation in bone marrow stromal cells by alendronate via ERK and JNK activation. Bone 2008. 43(1): p. 40-7.

327. Kaur G, Valarmathi MT, Potts JD, Jabbari E, Sabo-Attwood T, Wang Q. Regulation of osteogenic differentiation of rat bone marrow stromal cells on 2D nanorod substrates. Biomaterials 2010. 31(7): p. 1732-41.

328. Mauney JR, Blumberg J, Pirun M, Volloch V, Vunjak-Novakovic G, Kaplan DL. Osteogenic differentiation of human bone marrow stromal cells on partially demineralized bone scaffolds in vitro. Tissue Eng 2004. 10(1-2): p. 81-92.

329. Langer HF, Stellos K, Steingen C, et al. Platelet derived bFGF mediates vascular integrative mechanisms of mesenchymal stem cells in vitro. J Mol Cell Cardiol 2009. 47(2): p. 315-25.

330. Garcia-de-Alba C, Becerril C, Ruiz V, et al. Expression of matrix metalloproteases by fibrocytes: possible role in migration and homing. Am J Respir Crit Care Med 2010. 182(9): p. 1144-52.

331. Yagi H, Soto-Gutierrez A, Parekkadan B, et al. Mesenchymal stem cells: Mechanisms of immunomodulation and homing. Cell Transplant 2010. 19(6): p. 667-79.

332. Ratanavaraporn J, Furuya H, Kohara H, Tabata Y. Synergistic effects of the dual release of stromal cell-derived factor-1 and bone morphogenetic protein-2 from hydrogels on bone regeneration. Biomaterials 2011. 32(11): p. 2797-811.

333. Tsugawa J, Komaki M, Yoshida T, Nakahama K, Amagasa T, Morita I. Cell-printing and transfer technology applications for bone defects in mice. J Tissue Eng Regen Med 2011. 5(9): p. 695-703.

334. Wu C, Zhang Y, Zhou Y, Fan W, Xiao Y. A comparative study of mesoporous glass/silk and non-mesoporous glass/silk scaffolds: physiochemistry and in vivo osteogenesis. Acta Biomater 2011. 7(5): p. 2229-36.

335. Fan W, Crawford R, Xiao Y. Enhancing in vivo vascularized bone formation by cobalt chloride-treated bone marrow stromal cells in a tissue engineered periosteum model. Biomaterials 2010. 31(13): p. 3580-9.

336. Hinsenkamp M, Muylle L, Eastlund T, Fehily D, Noel L, Strong DM. Adverse reactions and events related to musculoskeletal allografts: reviewed by the World Health Organisation Project NOTIFY. Int Orthop 2012. 36(3): p. 633-41.

337. Herzog EL, Chai L, Krause DS. Plasticity of marrow-derived stem cells. Blood 2003. 102(10): p. 3483-93.

338. Wang Y, Volloch V, Pindrus MA, Blasioli DJ, Chen J, Kaplan DL. Murine osteoblasts regulate mesenchymal stem cells via WNT and cadherin pathways: mechanism depends on cell-cell contact mode. J Tissue Eng Regen Med 2007. 1(1): p. 39-50.

339. Bulnheim U, Muller P, Neumann HG, et al. Endothelial cells stimulate osteogenic differentiation of mesenchymal stem cells on calcium phosphate scaffolds. J Tissue Eng Regen Med 2012: p. doi: 10.1002/term.590.

340. F. LY, Wang S, Shi S. The role of recipient T cells in mesenchymal stem cell-based tissue regeneration. Int J Biochem Cell Biol 2012. 44(11): p. 2044-50.

341. Li F, Whyte N, Niyibizi C. Differentiating multipotent mesenchymal stromal cells generate factors that exert paracrine activities on exogenous MSCs: Implications for

Page 156: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

136 | P a g e

paracrine activities in bone regeneration. Biochem Biophys Res Commun 2012. 426(4): p. 475-9.

342. Langenbach F, Naujoks C, Kersten-Thiele PV, et al. Osteogenic differentiation influences stem cell migration out of scaffold-free microspheres. Tissue Eng Part A 2010. 16(2): p. 759-66.

343. Augello A, Kurth TB, De Bari C. Mesenchymal stem cells: a perspective from in vitro cultures to in vivo migration and niches. Eur Cell Mater 2010. 20: p. 121-33.

344. Kumar S, Wan C, Ramaswamy G, Clemens TL, Ponnazhagan S. Mesenchymal stem cells expressing osteogenic and angiogenic factors synergistically enhance bone formation in a mouse model of segmental bone defect. Mol Ther 2010. 18(5): p. 1026-34.

345. Gerhardt LC, Widdows KL, Erol MM, et al. The pro-angiogenic properties of multi-functional bioactive glass composite scaffolds. Biomaterials 2011. 32(17): p. 4096-108.

346. Willems WF, Larsen M, Giusti G, Friedrich PF, Bishop AT. Revascularization and bone remodeling of frozen allografts stimulated by intramedullary sustained delivery of FGF-2 and VEGF. J Orthop Res 2011. 29(9): p. 1431-6.

347. Jin T, Xu X, Hereld D. Chemotaxis, chemokine receptors and human disease. Cytokine 2008. 44(1): p. 1-8.

348. Maghazachi AA. Intracellular signaling events at the leading edge of migrating cells. Int J Biochem Cell Biol 2000. 32(9): p. 931-43.

349. Keeley EC, Mehrad B, Strieter RM. CXC chemokines in cancer angiogenesis and metastases. Adv Cancer Res 2010. 106: p. 91-111.

350. Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, Silberstein LE. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells 2006. 24(4): p. 1030-41.

351. Lee DH, Park BJ, Lee MS, et al. Chemotactic migration of human mesenchymal stem cells and MC3T3-E1 osteoblast-like cells induced by COS-7 cell line expressing rhBMP-7. Tissue Eng 2006. 12(6): p. 1577-86.

352. Tang JM, Wang JN, Zhang L, et al. VEGF/SDF-1 promotes cardiac stem cell mobilization and myocardial repair in the infarcted heart. Cardiovasc Res 2011. 91(3): p. 402-11.

353. Maes C, Kobayashi T, Selig MK, et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev Cell 2010. 19(2): p. 329-44.

354. Campbell JP, Karolak MR, Ma Y, et al. Stimulation of host bone marrow stromal cells by sympathetic nerves promotes breast cancer bone metastasis in mice. PLos Biol 2012. 10(7): p. e1001363.

355. Mareddy S, Dhaliwal N, Crawford R, Xiao Y. Stem cell-related gene expression in clonal populations of mesenchymal stromal cells from bone marrow. Tissue Eng Part A 2010. 16(2): p. 749-58.

356. Rahim S, Uren A. A real-time electrical impedance based technique to measure invasion of endothelial cell monolayer by cancer cells. J Vis Exp 2011(50).

357. Nam D, Mantell J, Bull D, Verkade P, Achim A. Active contour based segmentation for insulin granule cores in electron micrographs of beta islet cells. Conf Proc IEEE Eng Med Biol Soc 2012. 2012: p. 5339-42.

358. Wu C, Zhou Y, Xu M, et al. Copper-containing mesoporous bioactive glass scaffolds with multifunctional properties of angiogenesis capacity, osteostimulation and antibacterial activity. Biomaterials 2013. 34(2): p. 422-33.

Page 157: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

137 | P a g e

359. Gassmann M, Grenacher B, Rohde B, Vogel J. Quantifying Western blots: pitfalls of densitometry. Electrophoresis 2009. 30(11): p. 1845-55.

360. Ricart E. Current Status of Mesenchymal Stem Cell Therapy and Bone Marrow Transplantation in IBD. Dig Dis 2012. 30(4): p. 387-91.

361. Kon E, Filardo G, Roffi A, et al. Bone regeneration with mesenchymal stem cells. Clin Cases Miner Bone Metab 2012. 9(1): p. 24-7.

362. Cashman TJ, Gouon-Evans V, Costa KD. Mesenchymal stem cells for cardiac therapy: practical challenges and potential mechanisms. Stem Cell Rev 2012.

363. Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res 2008. 103(11): p. 1204-19.

364. Tammela T, Enholm B, Alitalo K, Paavonen K. The biology of vascular endothelial growth factors. Cardiovasc Res 2005. 65(3): p. 550-63.

365. Kanczler JM, Oreffo RO. Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cell Mater 2008. 15: p. 100-14.

366. Long Q, Liu W, Zhong J, et al. The tropism of neurally differentiated bone marrow stromal cells towards C6 glioma. Neurosci Lett 2011. 504(2): p. 135-40.

367. Ritter E, Perry A, Yu J, Wang T, Tang L, Bieberich E. Breast cancer cell-derived fibroblast growth factor 2 and vascular endothelial growth factor are chemoattractants for bone marrow stromal stem cells. Ann Surg 2008. 247(2): p. 310-4.

368. Schichor C, Birnbaum T, Etminan N, et al. Vascular endothelial growth factor A contributes to glioma-induced migration of human marrow stromal cells (hMSC). Exp Neurol 2006. 199(2): p. 301-10.

369. Sulpice E, Ding S, Muscatelli-Groux B, et al. Cross-talk between the VEGF-A and HGF signalling pathways in endothelial cells. Biol Cell 2009. 101(9): p. 525-39.

370. Coates TD, Watts RG, Hartman R, Howard TH. Relationship of F-actin distribution to development of polar shape in human polymorphonuclear neutrophils. J Cell Biol 1992. 117(4): p. 765-74.

371. Petrova TV, Makinen T, Alitalo K. Signaling via vascular endothelial growth factor receptors. Exp Cell Res 1999. 253(1): p. 117-30.

372. Kim DH, Yoo KH, Choi KS, et al. Gene expression profile of cytokine and growth factor during differentiation of bone marrow-derived mesenchymal stem cell. Cytokine 2005. 31(2): p. 119-26.

373. Bruns AF, Bao L, Walker JH, Ponnambalam S. VEGF-A-stimulated signalling in endothelial cells via a dual receptor tyrosine kinase system is dependent on co-ordinated trafficking and proteolysis. Biochem Soc Trans 2009. 37(Pt 6): p. 1193-7.

374. Kajdaniuk D, Marek B, Foltyn W, Kos-Kudla B. Vascular endothelial growth factor (VEGF) - part 1: in physiology and pathophysiology. Endokrynol Pol 2011. 62(5): p. 444-55.

375. Ruiz de Almodovar C, Fabre PJ, Knevels E, et al. VEGF mediates commissural axon chemoattraction through its receptor Flk1. Neuron 2011. 70(5): p. 966-78.

376. Cudmore MJ, Hewett PW, Ahmad S, et al. The role of heterodimerization between VEGFR-1 and VEGFR-2 in the regulation of endothelial cell homeostasis. Nat Commun 2012. 3: p. 972.

377. Doi Y, Yashiro M, Yamada N, Amano R, Noda S, Hirakawa K. VEGF-A/VEGFR-2 signaling plays an important role for the motility of pancreas cancer cells. Ann Surg Oncol 2012. 19(8): p. 2733-43.

378. Kranich S, Hattermann K, Specht A, Lucius R, Mentlein R. VEGFR-3/Flt-4 mediates proliferation and chemotaxis in glial precursor cells. Neurochem Int 2009. 55(8): p. 747-53.

Page 158: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

138 | P a g e

379. Fan W, Crawford R, Xiao Y. The ratio of VEGF/PEDF expression in bone marrow mesenchymal stem cells regulates neovascularization. Differentiation 2011. 81(3): p. 181-91.

380. Abid MR, Guo S, Minami T, et al. Vascular endothelial growth factor activates PI3K/Akt/forkhead signaling in endothelial cells. Arterioscler Thromb Vasc Biol 2004. 24(2): p. 294-300.

381. Chen J, Fang Y. A novel pathway regulating the mammalian target of rapamycin (mTOR) signaling. Biochem Pharmacol 2002. 64(7): p. 1071-7.

382. Taylor D, Hazenberg JG, Lee TC. Living with cracks: damage and repair in human bone. Nat Mater 2007. 6(4): p. 263-8.

383. De Long WG, Jr., Einhorn TA, Koval K, et al. Bone grafts and bone graft substitutes in orthopaedic trauma surgery. A critical analysis. J Bone Joint Surg Am 2007. 89(3): p. 649-58.

384. Sasso RC, LeHuec JC, Shaffrey C. Iliac crest bone graft donor site pain after anterior lumbar interbody fusion: a prospective patient satisfaction outcome assessment. J Spinal Disord Tech 2005. 18 Suppl: p. S77-81.

385. Ahlmann E, Patzakis M, Roidis N, Shepherd L, Holtom P. Comparison of anterior and posterior iliac crest bone grafts in terms of harvest-site morbidity and functional outcomes. J Bone Joint Surg Am 2002. 84-A(5): p. 716-20.

386. Giannoudis PV, Dinopoulos H, Tsiridis E. Bone substitutes: an update. Injury 2005. 36 Suppl 3: p. S20-7.

387. Ji W, Sun Y, Yang F, et al. Bioactive electrospun scaffolds delivering growth factors and genes for tissue engineering applications. Pharm Res 2011. 28(6): p. 1259-72.

388. Sabir MI, Xu X, Li L. A review on biodegradable polymeric materials for bone tissue engineering applications. J Mater Sci 2009. 44: p. 5713-24.

389. Liu B, Lun DX. Current application of beta-tricalcium phosphate composites in orthopaedics. Orthop Surg 2012. 4(3): p. 139-44.

390. Rahaman MN, Day DE, Bal BS, et al. Bioactive glass in tissue engineering. Acta Biomater 2011. 7(6): p. 2355-73.

391. Lee JTY, Wang K, Tsang WH, Chow KL. Comparative in vitro osteoinductivity study of CaP ceramics (HA, α-TCP, β-TCP) using 10T1/2 cells with different controls and possible correlations with other systems. J Biomater Nanobiotechnol 2011. 2: p. 163-72.

392. Garrido CA, Lobo SE, Turibio FM, Legeros RZ. Biphasic calcium phosphate bioceramics for orthopaedic reconstructions: clinical outcomes. Int J Biomater 2011. 2011: p. 129727.

393. Hench LL. The story of Bioglass. J Mater Sci Mater Med 2006. 17(11): p. 967-78. 394. Fu Q, Rahaman MN, Bal BS, Huang W, Day DE. Preparation and bioactive

characteristics of a porous 13-93 glass, and fabrication into the articulating surface of a proximal tibia. J Biomed Mater Res A 2007. 82(1): p. 222-9.

395. Vitale-Brovarone C, Ciapetti G, Leonardi E, et al. Resorbable glass-ceramic phosphate-based scaffolds for bone tissue engineering: synthesis, properties, and in vitro effects on human marrow stromal cells. J Biomater Appl 2011. 26(4): p. 465-89.

396. Fu Q, Rahaman MN, Bal BS, Bonewald LF, Kuroki K, Brown RF. Silicate, borosilicate, and borate bioactive glass scaffolds with controllable degradation rate for bone tissue engineering applications. II. In vitro and in vivo biological evaluation. J Biomed Mater Res A 2010. 95(1): p. 172-9.

397. Huang W, Day DE, Kittiratanapiboon K, Rahaman MN. Kinetics and mechanisms of the conversion of silicate (45S5), borate, and borosilicate glasses to hydroxyapatite in dilute phosphate solutions. J Mater Sci Mater Med 2006. 17(7): p. 583-96.

Page 159: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

139 | P a g e

398. Chen QZ, Thompson ID, Boccaccini AR. 45S5 Bioglass-derived glass-ceramic scaffolds for bone tissue engineering. Biomaterials 2006. 27(11): p. 2414-25.

399. Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polym Sci 2007. 32: p. 762-98.

400. Middleton JC, Tipton AJ. Synthetic biodegradable polymers as orthopedic devices. Biomaterials 2000. 21(23): p. 2335-46.

401. Rezwan K, Chen QZ, Blaker JJ, Boccaccini AR. Biodegradable and bioactive porous polymer/inorganic composite scaffolds for bone tissue engineering. Biomaterials 2006. 27(18): p. 3413-31.

402. Rich J, Jaakkola T, Tirri T, Narhi T, Yli-Urpo A, Seppala J. In vitro evaluation of poly(epsilon-caprolactone-co-DL-lactide)/ bioactive glass composites. Biomaterials 2002. 23(10): p. 2143-50.

403. Jain RA. The manufacturing techniques of various drug loaded biodegradable poly(lactide-co-glycolide) (PLGA) devices. Biomaterials 2000. 21(23): p. 2475-90.

404. Wang HW, Cheng CW, Li CW, Chang HW, Wu PH, Wang GJ. Fabrication of pillared PLGA microvessel scaffold using femtosecond laser ablation. Int J Nanomedicine 2012. 7: p. 1865-73.

405. Saito E, Liao EE, Hu WW, Krebsbach PH, Hollister SJ. Effects of designed PLLA and 50:50 PLGA scaffold architectures on bone formation in vivo. J Tissue Eng Regen Med 2013. 7(2): p. 99-111.

406. Han SH, Kim YH, Park MS, et al. Histological and biomechanical properties of regenerated articular cartilage using chondrogenic bone marrow stromal cells with a PLGA scaffold in vivo. J Biomed Mater Res A 2008. 87(4): p. 850-61.

407. Wahl DA, Czernuszka JT. Collagen-hydroxyapatite composites for hard tissue repair. Eur Cell Mater 2006. 11: p. 43-56.

408. Dawes E, Rushton N. The effects of lactic acid on PGE2 production by macrophages and human synovial fibroblasts: a possible explanation for problems associated with the degradation of poly(lactide) implants? Clin Mater 1994. 17(4): p. 157-63.

409. Athanasiou KA, Niederauer GG, Agrawal CM. Sterilization, toxicity, biocompatibility and clinical applications of polylactic acid/polyglycolic acid copolymers. Biomaterials 1996. 17(2): p. 93-102.

410. Hennink WE, van Nostrum CF. Novel crosslinking methods to design hydrogels. Adv Drug Deliv Rev 2002. 54(1): p. 13-36.

411. Patino MG, Neiders ME, Andreana S, Noble B, Cohen RE. Collagen: an overview. Implant Dent 2002. 11(3): p. 280-5.

412. Raman SS, Gopalakrishnan R, Wade RC, Subramanian V. Structural basis for the varying propensities of different amino acids to adopt the collagen conformation. J Phys Chem B 2011. 115(11): p. 2593-607.

413. Gelse K, Poschl E, Aigner T. Collagens--structure, function, and biosynthesis. Adv Drug Deliv Rev 2003. 55(12): p. 1531-46.

414. Glowacki J, Mizuno S. Collagen scaffolds for tissue engineering. Biopolymers 2008. 89(5): p. 338-44.

415. Ferreira AM, Gentile P, Chiono V, Ciardelli G. Collagen for bone tissue regeneration. Acta Biomater 2012. 8(9): p. 3191-200.

416. Miyata T, Taira T, Noishiki Y. Collagen engineering for biomaterial use. Clin Mater 1992. 9(3-4): p. 139-48.

417. Lynn AK, Yannas IV, Bonfield W. Antigenicity and immunogenicity of collagen. J Biomed Mater Res B Appl Biomater 2004. 71(2): p. 343-54.

418. Parikh SN. Bone graft substitutes in modern orthopedics. Orthopedics 2002. 25(11): p. 1301-9; quiz 10-1.

Page 160: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

140 | P a g e

419. Baumann LS, Kerdel F. The treatment of bovine collagen allergy with cyclosporin. Dermatol Surg 1999. 25(3): p. 247-9.

420. Kim IS, Lee EN, Cho TH, et al. Promising efficacy of Escherichia coli recombinant human bone morphogenetic protein-2 in collagen sponge for ectopic and orthotopic bone formation and comparison with mammalian cell recombinant human bone morphogenetic protein-2. Tissue Eng Part A 2011. 17(3-4): p. 337-48.

421. Lin L, Fu X, Zhang X, et al. Rat adipose-derived stromal cells expressing BMP4 induce ectopic bone formation in vitro and in vivo. Acta Pharmacol Sin 2006. 27(12): p. 1608-15.

422. Wei G, Jin Q, Giannobile WV, Ma PX. The enhancement of osteogenesis by nano-fibrous scaffolds incorporating rhBMP-7 nanospheres. Biomaterials 2007. 28(12): p. 2087-96.

423. Kim SS, Gwak SJ, Kim BS. Orthotopic bone formation by implantation of apatite-coated poly(lactide-co-glycolide)/hydroxyapatite composite particulates and bone morphogenetic protein-2. J Biomed Mater Res A. 2008. 87(1): p. 245-53.

424. Lau KH, Gysin R, Chen ST, Wergedal JE, Baylink DJ, Mohan S. Marrow stromal cell-based cyclooxygenase 2 ex vivo gene-transfer strategy surprisingly lacks bone-regeneration effects and suppresses the bone-regeneration action of bone morphogenetic protein 4 in a mouse critical-sized calvarial defect model. Calcif Tissue Int 2009. 85(4): p. 356-67.

425. Kinsella CR, Jr., Macisaac ZM, Cray JJ, et al. Novel animal model of calvarial defect: part III. Reconstruction of an irradiated wound with rhBMP-2. Plast Reconstr Surg 2012. 130(5): p. 643e-50e.

426. Forriol F, Longo UG, Concejo C, Ripalda P, Maffulli N, Denaro V. Platelet-rich plasma, rhOP-1 (rhBMP-7) and frozen rib allograft for the reconstruction of bony mandibular defects in sheep. A pilot experimental study. Injury 2009. 40 Suppl 3: p. S44-9.

427. Alonso N, Tanikawa DY, Freitas Rda S, Canan L, Jr., Ozawa TO, Rocha DL. Evaluation of maxillary alveolar reconstruction using a resorbable collagen sponge with recombinant human bone morphogenetic protein-2 in cleft lip and palate patients. Tissue Eng Part C Methods 2010. 16(5): p. 1183-9.

428. Lee KB, Taghavi CE, Murray SS, Song KJ, Keorochana G, Wang JC. BMP induced inflammation: a comparison of rhBMP-7 and rhBMP-2. J Orthop Res 2012. 30(12): p. 1985-94.

429. Deutsch H. High-dose bone morphogenetic protein-induced ectopic abdomen bone growth. Spine J 2010. 10(2): p. e1-4.

430. Benglis D, Wang MY, Levi AD. A comprehensive review of the safety profile of bone morphogenetic protein in spine surgery. Neurosurgery 2008. 62(5 Suppl 2): p. ONS423-31; discussion ONS31.

431. Peng H, Wright V, Usas A, et al. Synergistic enhancement of bone formation and healing by stem cell-expressed VEGF and bone morphogenetic protein-4. J Clin Invest 2002. 110(6): p. 751-9.

432. Kanczler JM, Ginty PJ, White L, et al. The effect of the delivery of vascular endothelial growth factor and bone morphogenic protein-2 to osteoprogenitor cell populations on bone formation. Biomaterials 2010. 31(6): p. 1242-50.

433. Huang YC, Kaigler D, Rice KG, Krebsbach PH, Mooney DJ. Combined angiogenic and osteogenic factor delivery enhances bone marrow stromal cell-driven bone regeneration. J Bone Miner Res 2005. 20(5): p. 848-57.

434. Lo DD, Hyun JS, Chung MT, et al. Repair of a critical-sized calvarial defect model using adipose-derived stromal cells harvested from lipoaspirate. J Vis Exp 2012(68).

Page 161: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

141 | P a g e

435. Dupont KM, Sharma K, Stevens HY, Boerckel JD, Garcia AJ, Guldberg RE. Human stem cell delivery for treatment of large segmental bone defects. Proc Natl Acad Sci U S A 2010. 107(8): p. 3305-10.

436. Chanda D, Kumar S, Ponnazhagan S. Therapeutic potential of adult bone marrow-derived mesenchymal stem cells in diseases of the skeleton. J Cell Biochem 2010. 111(2): p. 249-57.

437. Macfarlane RJ, Graham SM, Davies PS, et al. Anti-inflammatory role and immunomodulation of mesenchymal stem cells in systemic joint diseases: potential for treatment. Expert Opin Ther Targets 2013. 17(3): p. 243-54.

438. Kuo YR, Chen CC, Goto S, et al. Immunomodulatory effects of bone marrow-derived mesenchymal stem cells in a swine hemi-facial allotransplantation model. PLoS One 2012. 7(4): p. e35459.

439. Mokbel AN, El Tookhy OS, Shamaa AA, Rashed LA, Sabry D, El Sayed AM. Homing and reparative effect of intra-articular injection of autologus mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord 2011. 12: p. 259.

440. Sordi V. Mesenchymal stem cell homing capacity. Transplantation 2009. 87(9 Suppl): p. S42-5.

441. Meyerrose T, Olson S, Pontow S, et al. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev 2010. 62(12): p. 1167-74.

442. Otsuru S, Tamai K, Yamazaki T, Yoshikawa H, Kaneda Y. Circulating bone marrow-derived osteoblast progenitor cells are recruited to the bone-forming site by the CXCR4/stromal cell-derived factor-1 pathway. Stem Cells 2008. 26(1): p. 223-34.

443. Kitaori T, Ito H, Schwarz EM, et al. Stromal cell-derived factor 1/CXCR4 signaling is critical for the recruitment of mesenchymal stem cells to the fracture site during skeletal repair in a mouse model. Arthritis Rheum 2009. 60(3): p. 813-23.

444. Colnot C. Cell sources for bone tissue engineering: insights from basic science. Tissue Eng Part B Rev 2011. 17(6): p. 449-57.

445. Xia Z, Locklin RM, Triffitt JT. Fates and osteogenic differentiation potential of human mesenchymal stem cells in immunocompromised mice. Eur J Cell Biol 2008. 87(6): p. 353-64.

446. Zong C, Xue D, Yuan W, et al. Reconstruction of rat calvarial defects with human mesenchymal stem cells and osteoblast-like cells in poly-lactic-co-glycolic acid scaffolds. Eur Cell Mater 2010. 20: p. 109-20.

447. Tasso R, Augello A, Boccardo S, et al. Recruitment of a host's osteoprogenitor cells using exogenous mesenchymal stem cells seeded on porous ceramic. Tissue Eng Part A 2009. 15(8): p. 2203-12.

448. Le Blanc K, Gotherstrom C, Ringden O, et al. Fetal mesenchymal stem-cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation 2005. 79(11): p. 1607-14.

449. Guillot PV, Abass O, Bassett JH, et al. Intrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis imperfecta mice. Blood 2008. 111(3): p. 1717-25.

450. Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am 2005. 87(7): p. 1430-7.

451. Rodrigues MT, Lee BK, Lee SJ, et al. The effect of differentiation stage of amniotic fluid stem cells on bone regeneration. Biomaterials 2012. 33(26): p. 6069-78.

452. Kim HP, Ji YH, Rhee SC, Dhong ES, Park SH, Yoon ES. Enhancement of bone regeneration using osteogenic-induced adipose-derived stem cells combined with

Page 162: Interactions between Undifferentiated and Osteogenically ...eprints.qut.edu.au/63002/1/Yinghong_Zhou_Thesis.pdf · Book chapter in Methods in Molecular Biology, 2013. Springer. Accepted

142 | P a g e

demineralized bone matrix in a rat critically-sized calvarial defect model. Curr Stem Cell Res Ther 2012. 7(3): p. 165-72.

453. Yang P, Wang C, Shi Z, et al. Prefabrication of vascularized porous three-dimensional scaffold induced from rhVEGF(165): a preliminary study in rats. Cells Tissues Organs 2009. 189(5): p. 327-37.

454. Patel ZS, Young S, Tabata Y, Jansen JA, Wong ME, Mikos AG. Dual delivery of an angiogenic and an osteogenic growth factor for bone regeneration in a critical size defect model. Bone 2008. 43(5): p. 931-40.

455. Seebach C, Henrich D, Wilhelm K, Barker JH, Marzi I. Endothelial progenitor cells improve directly and indirectly early vascularization of mesenchymal stem cell-driven bone regeneration in a critical bone defect in rats. Cell Transplant 2012. 21(8): p. 1667-77.

456. Horner EA, Kirkham J, Wood D, et al. Long bone defect models for tissue engineering applications: criteria for choice. Tissue Eng Part B Rev 2010. 16(2): p. 263-71.

457. Jones E, Yang X. Mesenchymal stem cells and bone regeneration: current status. Injury 2011. 42(6): p. 562-8.

458. Scott MA, Levi B, Askarinam A, et al. Brief review of models of ectopic bone formation. Stem Cells Dev 2012. 21(5): p. 655-67.

459. Montero JA, Kilian B, Chan J, Bayliss PE, Heisenberg CP. Phosphoinositide 3-kinase is required for process outgrowth and cell polarization of gastrulating mesendodermal cells. Curr Biol 2003. 13(15): p. 1279-89.

460. Inoue T, Meyer T. Synthetic activation of endogenous PI3K and Rac identifies an AND-gate switch for cell polarization and migration. PLoS One 2008. 3(8): p. e3068.