190
DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE SCAFFOLDS FOR BONE REGENERATION Jiongyu Ren BEng, MEng Submitted in fulfilment of the requirements for the degree of Doctor of Philosophy Institute of Health and Biomedical Innovation Faculty of Science and Engineering Queensland University of Technology 2017

DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE SCAFFOLDS FOR BONE

REGENERATION

Jiongyu Ren BEng, MEng

Submitted in fulfilment of the requirements for the degree of

Doctor of Philosophy

Institute of Health and Biomedical Innovation

Faculty of Science and Engineering

Queensland University of Technology

2017

Page 2: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 3: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration i

Keywords

Melt-electrospinning, hybrid melt-electrospinning, bioactive glass,

polycaprolactone, composite scaffold, bone tissue engineering, strontium-substituted

bioactive glass, bone histology

Page 4: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

ii development of melt electrospun composite scaffolds for bone regeneration

Abstract

A scaffold based approach of tissue engineering can potentially circumvent the

issues associated with current bone grafting treatment for bone defect regeneration.

Polycaprolactone (PCL) is a resorbable polymer used extensively in bone tissue

engineering owing to good structural properties and processability. Strontium-

substituted bioactive glass (SrBG) has the ability to promote osteogenesis and may be

incorporated into scaffolds intended for bone repair. The incorporation of SrBG

particle filler phase into PCL matrix produced bioactive PCL/SrBG composite that

possesses good processability. Melt-electrospinning is an additive manufacturing

technique to produce porous scaffolds with high surface area to volume ratio. In this

PhD project, I contributed to three key processes of scaffold development including

scaffold fabrication, in vitro assessment of scaffolds and the optimization of endpoint

histological techniques for the ex vivo analysis of these scaffolds. In the first study,

PCL/SrBG (10 wt%) composite scaffolds were developed using the technique of melt-

electrospinning. The characterization of these scaffolds demonstrated their bioactivity

and favourable in vitro characteristics. Osteoblast-precursor cells cultured on

PCL/SrBG (10 wt%) scaffolds showed improved osteogenic properties indicated by

enhanced alkaline phosphatase (ALP) activity when compared to scaffolds made of

pure PCL. In order to enhance osteoblastic differentiation by the scaffolds, I increased

the percentage of SrBG particles up to 50 wt% in the PCL matrix in study 2. This was

made possible by a novel hybrid melt-electrospinning technique that was developed to

produce scaffolds with controlled fibre spacing and lay down pattern. The PCL/SrBG

(50 wt%) scaffolds showed significantly enhanced bioactivity compared to PCL/SrBG

(10 wt%) ones. In vitro assessment of PCL/SrBG (50 wt%) scaffolds indicated these

scaffolds were osteogenic in vitro as evidenced by the significantly enhanced alkaline

phosphatase activity compared to PCL control scaffolds. While the first and second

study were carried out to improve the fabrication of bioactive scaffolds as suitable

bone defect implants, the third study of this Ph.D. project was carried out in parallel to

improve the histological tools for the evaluation of future animal implantation

experiments with these PCL/SrBG composite scaffolds. Using samples from different

species used in typical biomaterial implantation experiments found in the literature, I

optimised all the essential techniques, resulting in detailed protocols for the

Page 5: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration iii

standardized histological processing of animal native bone and tissue engineered bone

explants.

Page 6: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 7: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration i

List of Publications

1. A Berner, M A Woodruff, C X F Lam, M T Arafat, S Saifzadeh, R Steck, J

Ren, M Nerlich, A K Ekaputra, I Gibson, D W Hutmacher, Biomimetic tubular

nanofiber mesh and platelet rich plasma-mediated delivery of growth factors for large

bone defect regeneration, Cell and Tissue Research, 2012

2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J Ren, C Vaquette, J Qiyi

Zhang, M Nerlich, R E Guldberg, D W Hutmacher, M A Woodruff, Effects of

scaffolds architecture on bone healing, International Journal of Oral and Maxillofacial

Surgery, 2013

3. Jiongyu Ren, Keith A Blackwood, Amir Doustgani, Patrina P Poh, Roland

Steck, Molly M Stevens, Maria A Woodruff, Melt-electrospun polycaprolactone

strontium-substituted bioactive glass scaffolds for bone regeneration, Journal of

Biomedical Materials Research Part A, 2014

4. Nikola Ristovski, Nathalie Bock, Sam Liao, Sean K Powell, Jiongyu

Ren, Giles T S Kirby, Keith A Blackwood, Maria A Woodruff, Improved fabrication

of melt electrospun tissue engineering scaffolds using direct writing and advanced

electric field contorl, Biointerphases, 2015

5. K. A. Blackwood, N. Ristovski, S. Liao, N. Bock, J. Ren, G. T. S. Kirby, M.

M. Stevens, R. Steck, M. A. Woodruff, Improving Electrospun Fibre Stacking with

Direct Writing for Developing Scaffolds for Tissue Engineering for Non-load Bearing

Bone, IFMBE Proceedings, 2015

Papers in preparation

Jiongyu Ren, Keith A Blackwood, Seamus Tredinnick, Roland Steck, Giles T Kirby,

Christina Theodoropoulos, Flavia M Savi, Maria A Woodruff, Development of

optimised histological processes for analysis of large and complex bone and implants

Jiongyu Ren, Keith A Blackwood, Roland Steck, Molly M Stevens, Maria A

Woodruff, Developing Strontium-substituted Bioactive glass and Polycaprolactone

composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Page 8: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

ii development of melt electrospun composite scaffolds for bone regeneration

Table of Contents

Keywords .................................................................................................................................. i

Abstract .................................................................................................................................... ii

List of Publications .................................................................................................................... i

Papers in preparation ................................................................................................................. i

Table of Contents ..................................................................................................................... ii

List of Figures .......................................................................................................................... v

List of Tables ......................................................................................................................... viii

List of Abbreviations ............................................................................................................... ix

Statement of Original Authorship ........................................................................................... xi

Acknowledgements ................................................................................................................ xii

Chapter 1: Introduction ...................................................................................... 1

1.1 Overview ........................................................................................................................ 1

1.2 Purpose of research ........................................................................................................ 4

1.3 Significance of research ................................................................................................. 5

1.4 Thesis Outline ................................................................................................................ 6

Chapter 2: Research Hypothesis and Aims ....................................................... 9

2.1 Hypothesis ...................................................................................................................... 9

2.2 Research Aims ............................................................................................................... 9

Chapter 3: Literature review ............................................................................ 15

3.1 Bone Biology ............................................................................................................... 15 3.1.1 Structure of bone ................................................................................................ 15 3.1.2 Bone fracture healing ......................................................................................... 18

3.2 Current treatments for large bone defects .................................................................... 19 3.2.1 Bone grafting ..................................................................................................... 19

3.3 Bone Tissue Engineering ............................................................................................. 20 3.3.1 3D printing scaffolds ......................................................................................... 21 3.3.2 Tissue engineering scaffolds .............................................................................. 21 3.3.3 TE scaffolds specifications ................................................................................ 22

3.4 Scaffold materials ........................................................................................................ 23 3.4.1 Polymers ............................................................................................................ 23 3.4.2 Bioactive glass (BG) .......................................................................................... 25 3.4.3 Biodegradable polymer/bioactive glass composites .......................................... 28

3.5 Scaffold fabrication techniques .................................................................................... 30 3.5.1 History of electrospinning ................................................................................. 32 3.5.2 Basic electrospinning equipment ....................................................................... 32 3.5.3 Theory of electrospinning .................................................................................. 33 3.5.4 Process of electrospinning ................................................................................. 34 3.5.5 Melt-electrospinning .......................................................................................... 35 3.5.6 Hybrid electrospinning system .......................................................................... 36

Page 9: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration iii

3.6 TE scaffolds assessment ...............................................................................................37 3.6.1 Histology ............................................................................................................38

3.7 Conclusion ....................................................................................................................42

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration ...................................... 45

4.1 Introduction ..................................................................................................................45

4.2 Materials and Methods .................................................................................................47 4.2.1 Scaffold fabrication and characterisation ...........................................................47 4.2.2 Ion dissolution and precipitation ........................................................................48 4.2.3 In vitro studies ....................................................................................................49 4.2.4 Statistical analyses ..............................................................................................54

4.3 Results ..........................................................................................................................54 4.3.1 Characterization of PCL and PCL/SrBG scaffolds ............................................54 4.3.2 Ion dissolution and precipitation analysis ..........................................................55 4.3.3 In vitro studies ....................................................................................................57

4.4 Discussion .....................................................................................................................64

4.5 Conclusions of study 1..................................................................................................71

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode ............................................................... 73

5.1 Introduction ..................................................................................................................73

5.2 Materials and Methods .................................................................................................75 5.2.1 PCL/SrBG composite preparation ......................................................................75 5.2.2 Scaffold fabrication ............................................................................................76 5.2.3 Scaffold characterisation ....................................................................................77 5.2.4 In vitro studies ....................................................................................................77 5.2.5 Statistical analyses ..............................................................................................81

5.3 Results ..........................................................................................................................82 5.3.1 Particle grinding and sizing ................................................................................82 5.3.2 Scaffold fabrication ............................................................................................82 5.3.3 Characterisation ..................................................................................................83 5.3.4 In vitro studies ....................................................................................................89

5.4 Discussion .....................................................................................................................94

5.5 Conclusion of study 2 .................................................................................................102

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants ............................................... 105

6.1 Introduction ................................................................................................................105

6.2 Materials and Methods ...............................................................................................108 6.2.1 Bone tissues and pre-processing preparation....................................................108 6.2.2 Processing and embedding ...............................................................................109 6.2.3 Sectioning .........................................................................................................111 6.2.4 Staining .............................................................................................................114 6.2.5 Microscopy and image documentation .............................................................119

6.3 Results and Discussion ...............................................................................................119 6.3.1 Study Overview ................................................................................................119 6.3.2 Stain optimization and comparison ..................................................................120

Page 10: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

iv development of melt electrospun composite scaffolds for bone regeneration

6.4 Conclusion of study 3 ................................................................................................ 133

6.5 Supplementary Figures .............................................................................................. 137

Chapter 7: Conclusions.................................................................................... 141

7.1 Research Summary .................................................................................................... 141

7.2 Summary of Chapter 4 (Study 1) ............................................................................... 142

7.3 Summary of Chapter 5 (study 2) ................................................................................ 143

7.4 Summary of Chapter 6 (Study 3) ............................................................................... 144

7.5 Limitations and recommendation for future work ..................................................... 145 7.5.1 Composite scaffold design ............................................................................... 145 7.5.2 In vivo investigation of composite scaffolds .................................................... 146

7.6 Concluding Remarks .................................................................................................. 148

References ............................................................................................................... 151

Appendices .............................................................................................................. 169

Page 11: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration v

List of Figures

Figure 1.1 General development process of bone TE scaffolds................................... 3

Figure 1.2 Overview of PCL/SrBG scaffolds development in this PhD project. ........ 4

Figure 3.1 The seven hierarchy levels of bone structure, as demonstrated by Weiner and Wagner [18]. ............................................................................. 16

Figure 3.2 Schematic illustration of long bone and its micro-structure [23] ............. 17

Figure 3.3 The four stages of bone fracture regeneration [24] .................................. 19

Figure 3.4 Schematic overview of the scaffold-based approach to bone TE [12] ..... 22

Figure 3.5 The degradation and elimination pathway of PCL. .................................. 25

Figure 3.6 The effect of bioactive glass ion dissolution products on biological responses [49] .............................................................................................. 27

Figure 3.7 Schematic illustration of typical electrospinning setup (not to scale) [82] ............................................................................................................... 33

Figure 3.8 A schematic summary of electrospinning configurations for desired fibre alignments [87] .................................................................................... 34

Figure 4.1 Electrospun scaffolds light microscopy and microCT characterization. ........................................................................................... 55

Figure 4.2 EDX image of melt-electrospun scaffolds after 14 days incubation in α-MEM. ................................................................................................... 57

Figure 4.3 LIVE/DEAD staining of MC3T3 cells cultured on melt-electrospun scaffolds. ...................................................................................................... 58

Figure 4.4 SEM images of MC3T3 cells cultured on PCL (a)(b) and (c) and PCL/SrBG (d)(e) and (f) scaffolds. .............................................................. 59

Figure 4.5 Confocal laser scanning microscopy images of MC3T3 cells cultured on melt-electrospun PCL/SrBG (a) and PCL scaffolds (b) for 3 days. .......................................................................................................... 59

Figure 4.6 MTT metabolic activity assay of MC3T3 cells over 28 days culture....... 60

Figure 4.7 Normalised ALP activity of MC3T3 cells cultured on melt-electrospun PCL and PCL/SrBG scaffolds cultured in osteogenic and control media. .............................................................................................. 61

Figure 4.8 Alizarin red S staining of PCL and PCL/SrBG scaffolds cultured with MC3T3 cells in control and osteogenic media over 28 days. .............. 62

Figure 4.9 Gene expression of osteoblast markers ALP (a) and OCN (b), in all experimental groups over 28 days. .............................................................. 63

Figure 4.10 Van Gieson staining of PCL and PCL/SrBG scaffolds cultured with MC3T3 cells in control and osteogenic media over 28 days. .............. 64

Figure 5.1 SrBG particle size distribution before grinding, after grinding and after drying. .................................................................................................. 82

Page 12: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

vi development of melt electrospun composite scaffolds for bone regeneration

Figure 5.2 SEM images of 50% PCL/SrBG (a-d) and PCL (e-h) scaffolds. .............. 84

Figure 5.3 Confocal laser scanning microscopy images of PCL/SrBG scaffolds stained by Alizarin red S. ............................................................................. 85

Figure 5.4 Backscattered SEM image of a PCL/SrBG scaffold. ............................... 86

Figure 5.5 Elemental concentrations of Ca2+ (a), PO43- (b), Si4+ (c), and Sr2+ (d)

of a-MEM media incubated with 50% PCL/SrBG and PCL scaffolds over 28 days as determined by ICP-MS testing. .......................................... 87

Figure 5.6 Example surface elemental compositions of 50% PCL/SrBG scaffolds after 3h (a), 6h (b), 1 day (c) and 2 days (d) in a-MEM media as determined by EDX. ..................................................................... 88

Figure 5.7 LIVE/DEAD staining of MC3T3 cells cultured on melt-electrospun scaffolds. ...................................................................................................... 90

Figure 5.8 SEM images of MC3T3 cells cultured on PCL (a) and (b) and PCL/SrBG (c) and (d) scaffolds. .................................................................. 91

Figure 5.9 Confocal laser scanning microscopy images of MC3T3 cells cultured on melt-electrospun ........................................................................ 91

Figure 5.10 MTT metabolic activity assay of MC3T3 cells over 28 days culture. .......................................................................................................... 92

Figure 5.11 Normalised ALP activity of MC3T3 cells cultured on melt-electrospun PCL and PCL/SrBG scaffolds cultured in osteogenic and control media. ............................................................................................... 93

Figure 5.12 Gene expression of osteoblast markers ALP (a) and OPN (b) as fold change to PCL control group in all experimental groups over 28 days. ............................................................................................................. 94

Figure 5.13 The direct comparison of Si4+ and Sr2+ ion concentration of PCL/SrBG (10wt%) scaffolds in study 1 and PCL/SrBG (50wt%) scaffolds in study 2. ................................................................................... 103

Figure 6.1 Schematic summary of study 3 design. .................................................. 109

Figure 6.2 An overview of resin block preparation and ground sectioning process. ....................................................................................................... 113

Figure 6.3 Comprehensive comparison of Goldner’s trichrome staining on resin ground sections and paraffin sections of bone of all four animal species. ....................................................................................................... 121

Figure 6.4 Specimen preparation and Goldner’s trichrome staining results of a tissue engineered sheep tibia with a 3 cm critical sized defect post mechanical testing. ..................................................................................... 125

Figure 6.5 Specimen preparation and staining results of sheep tibiae with scaffolds implanted into 3 cm critical sized defects. ................................. 129

Figure 6.6 Comparison of mouse paws prepared as resin ground sections, resin thin sections and paraffin sections stained with Goldner’s trichrome stain. ........................................................................................................... 130

Page 13: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration vii

Figure 6.7 Overview and high magnification images of a whole mouse hind paw and a sheep tibia/femur with porous titanium implant prepared by ground sectioning and stained with Goldner’s trichrome. ......................... 132

Figure A.1 Goldner’s trichrome staining of resin embedded sheep tibia bone tissues around titanium implant, sections obtained by ground sectioning technique................................................................................... 169

Figure A.2 Goldner’s trichrome staining of resin embedded sheep tibia native bone tissues and regenerated bone tissues around implanted PCL scaffolds ..................................................................................................... 170

Figure A.3 Goldner’s trichrome staining of resin embedded sheep tibia bone tissues around Ti implant, sections obtained by ground sectioning technique [132]. ......................................................................................... 171

Page 14: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

viii development of melt electrospun composite scaffolds for bone regeneration

List of Tables

Table 3.1 Properties of biodegradable polymers as bone scaffold materials [35,36] .......................................................................................................... 24

Table 3.2 Reaction stages of HCA layer formation on 45S5 BG [54] ....................... 26

Table 3.3 Comparison of different bone scaffold fabrication techniques [50,66] ..... 31

Table 3.4 Important parameters for electrospinning process [80] ............................. 34

Table 4.1 Parameters for melt-electrospun scaffold fabrication ................................ 48

Table 4.2 In vitro experimental groups of study 1. PCL/SrBG scaffolds (10 wt%) were studied in both growth media and osteogenic media and PCL scaffolds were studied as control. ........................................................ 49

Table 5.1 Parameters for 50 wt% PCL/SrBG and PCL scaffolds fabrication ............ 76

Table 5.2 In vitro experimental groups of study 2. PCL/SrBG scaffolds (50 wt%) were studied in both growth media and osteogenic media and PCL scaffolds were studied as control. ........................................................ 78

Table 5.3 Web of Science search results of ‘melt-electrospinning’ AND ‘polymer composite’ on 10/10/2016 ............................................................ 95

Table 6.1 Summary of embedding media and sectioning techniques in study 3. .... 107

Table 6.2 Preparation of Technovit 9100 New® solutions ....................................... 111

Table 6.3 Suitable sandpaper grits according to section thickness: a. final thickness=27 μm; b. final thickness=50 μm .............................................. 114

Table 6.4 Goldner’s trichrome staining solutions .................................................... 115

Table 6.5 Von Kossa/MacNeal’ tetrachrome staining solutions .............................. 117

Table 6.6 Summary of the advantages and disadvantages of commonly used media for histology embedding: paraffin, MMA resin and Technovit 9100 resin. .................................................................................................. 134

Table 6.7 Summary of the advantages and disadvantages of commonly used sectioning techniques for resin embedded specimens ................................ 136

Page 15: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration ix

List of Abbreviations

Full name Abbreviation 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide MTT Additive manufacturing AM Alkaline phosphatase ALP Alpha-minimum essential medium α-MEM Bioactive glass BG Bone morphogenetic protein BMP Bovine serum albumin BSA Calcium Phosphate CaP Confocal laser scanning microscopy CLSM Dimethyl sulfoxide DMSO Double-stranded deoxyribonucleic acid dsDNA Energy-dispersive X-ray spectroscopy EDX Electron probe microanalyzer EPMA Backscattered electron BSE Extracellular matrix ECM Fetal bovine serum FBS Hydroxyapatite HA Inductively coupled plasma - optical emission spectrometer ICP-OES Inductively coupled plasma - mass spectrometer ICP-MS Mega Pascal Mpa Micro-computed tomography μCT Nano-sized bioactive glass nBG Osteocalcin OCN Osteopontin OPN Power of hydrogen pH Phosphate buffer solution PBS Poly(3-hydroxybutyarate) P(3HB) Poly(caprolactone-co-DL_lactide) P(CL-DLLA) Poly(lactic acid) PLA Poly(glycolic acid) PGA Poly(L-lactic-co-glycolic acid) PLGA Poly(L-lactide) PLLA Poly(trimethylene carbonate) TMC Polycaprolactone PCL Protein kinase PK Rapid prototyping RP Real-time-quantitative polymerase chain reaction RT-qPCR Scanning electron microscopy SEM Sodium hydroxide NaOH Strontium-substituted bioactive glass SrBG Hydroxycarbonate apatite HCA Three dimensional 3D Tissue engineering TE

Page 16: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

x development of melt electrospun composite scaffolds for bone regeneration

Methyl methacrylate MMA Hematoxylin & Eosin H & E Von Willebrand factor vWF

Page 17: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

development of melt electrospun composite scaffolds for bone regeneration xi

Statement of Original Authorship

The work contained in this thesis has not been previously submitted to meet

requirements for an award at this or any other higher education institution. To the best

of my knowledge and belief, the thesis contains no material previously published or

written by another person except where due reference is made.

Signature: QUT Verified Signature

Date: May 2017

Page 18: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

xii development of melt electrospun composite scaffolds for bone regeneration

Acknowledgements

First and foremost I want to thank my supervisors, Associate Professor Maria A.

Woodruff, Dr. Roland Steck and Dr. Keith A. Blackwood. I could not have achieved

this major milestone in my career without their ongoing guidance and support. I am

especially grateful to A/Prof Woodruff for supporting me not only by providing a

research assistantship, but also academically through the rough road to finish this PhD

thesis. I acknowledge the professional help in editing this thesis from my supervisors

Dr. Roland Steck and A/Prof Mia Woodruff.

This thesis is also the result of support and help from dozens of remarkable

individuals at QUT. I would like to first thank my colleagues of the Biofabrication &

Tissue Morphology group and the Regenerative Medicine group who have contributed

to my PhD project. In particular, I thank Dr. Patrina Poh for sharing the knowledge

and techniques of in vitro assays and the support from CARF Histology Facility.

I acknowledge the following colleagues for their consultation: Dr

Henrietta Cathey (EPMA), Mr Tony Raftery (micronizing mill), Mr David Page

(Malvern Mastersizer), Emeritus Professor Graeme George and Dr John Colwell

(SrBG surface modification), Dr Charlotte Allen, Dr Sunny Hu and Mr. Mitchell De

Bruyn (ICP-MS), Dr Marie-Luise Wille (μCT), Dr Tong Li (AFM), Ms Rachel

Hancock (SEM), Dr. Christina Theodoropoulos and Dr Leonore de Boer (CLSM) and

Mr Shane Russell (ICP-OES).

I would like to acknowledge our collaborator professor Molly M. Stevens and

Dr. Anu Solanki from Imperial College London for providing bioactive glass that

made this project possible. Furthermore, I acknowledge Australian Research Council

for sponsoring this project (grant LP110200082 and LP100200084), and QUT APA

scholarship for the financial support.

Last but not least, I thank the support from my parents, parents-in-law, my wife

Xue Zhang, my daughter Ravenna Zirong Ren and my son Joshua Zitao Ren, I dedicate

this PhD thesis to you.

Page 19: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 1: Introduction 1

Chapter 1: Introduction

1.1 OVERVIEW

Musculoskeletal disorders have a significant socioeconomic impact worldwide.

They place a huge burden on the healthcare systems and community. Critical-sized

bone defects (a defect size which will not naturally heal) are one of the biggest

contributors, among these only a few patients will fully restore their limb function to

the level whereby they can return to previous employment [1]. In Australia, millions

of people are suffering from severe pain and disability caused by musculoskeletal

conditions and fatalities still occur due to ineffective available treatments. According

to Authoritative information and statistics to promote better health and wellbeing

(AIHW), 6.1 million Australians are affected by musculoskeletal conditions and

725,500 Australians are diagnosed with osteoporosis in 2011 to 2012 [2]. As a result,

musculoskeletal conditions have become a major burden to direct health expenditure

which accounted for $9153.7 million in 2012 [3], the fourth leading contributor

following cardiovascular diseases, oral health and mental disorder [4]. The figures

from the United States are even higher, reaching US$849 billion [5]. Besides the

economic expenditures, musculoskeletal conditions also affect the lives of patients and

place a significant burden on the community and health care services. Therefore,

researchers are actively seeking effective treatment for large bone defects in clinical

practice.

Current clinical treatments for critical bone defects rely on bone grafting,

including autografting (transplanting a patient’s own bone), allografting and

xenografting (transplanting bone from a donor or another species, respectively).

Although these procedures exhibited healing capacities, the bone regeneration results

have not always been satisfactory and these techniques have shown prominent

disadvantages such as disease transmission, immunogenic response, and

complications in bone harvesting surgeries. These complications have driven the

development of biomaterials which may ultimately substitute and improve on current

techniques. Over the last two decades, many bone substitute materials have been

evaluated with the intention of replacing autografts or allografts. The endeavours of

Page 20: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

2 Chapter 1: Introduction

researchers over the years led to the emergence of a new field of research known as

Tissue Engineering (TE).

TE combines the knowledge of both engineering and the life sciences with the

aim to produce tissue-like substitutes for the purpose of restoring or improving the

natural tissue function. Researchers in the bone TE field have extensively investigated

using polymeric scaffolds as potential bone graft substitutes. Amongst the variety of

clinical grade scaffolds available to patients today, bioresorbable polymeric scaffolds

have drawn a lot of attention, as they have the potential to integrate with the host bone

and degrade away within a controllable time frame to be replaced by newly-formed

bone tissue. However, current research suggests that no single component material can

satisfy all the requirements of viable bone-substituting scaffolds, due to the large

number of characteristics required for a suitable tissue engineered material. Composite

materials composed of polymers and bioactive glass/ceramic particles have emerged

as highly promising candidates because the bulk materials combine the processability

of polymers with the bioactivity of particle filler phase. The fine particle filler phase

also enhances the mechanical stability of polymer matrix making the composite

material stronger than polymer only. Composite scaffolds of high porosity (>80%) are

rarely reported for the treatment of critical bone defects within medium to high load-

bearing sites, owing to a lack of strength and the fact that they are still in a developing

stage. When developing a tissue engineered scaffold for bone regeneration we need to

consider the properties of the materials. Polycaprolactone (PCL) has good mechanical

properties and processibility; strontium-substituted bioactive glass (SrBG) has the

ability to promote osteogenesis. The PCL/SrBG composite combines the advantages

of these two materials and therefore is superior to PCL or SrBG only.

Several tissue engineering approaches have been developed to fabricate porous

3D composite scaffolds with high pore interconnectivity. Among them,

electrospinning has recently gained substantial research attention. Electrospinning is a

versatile technique for the production of fibres ranging from the micro to nano scale

in diameter, and the scaffolds produced with these micro- or nanofibres are of

extremely high surface area to volume ratio. This technique is well established in the

field of tissue engineering due to the ease to set up and low cost to run. Many different

polymers can be spun into scaffolds with interconnective pores using electrospinning.

There are mainly two ways of preparing polymers for electrospinning: dissolving the

Page 21: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 1: Introduction 3

polymer in an organic solvent (solution- electrospinning) and melting the polymer

(melt-electrospinning). Melt- electrospinning overcomes technical restrictions of

solution-electrospinning governed by solvent accumulation and toxicity. In spite of the

potential benefits of melt spinning, it has gained less attention in the TE field compared

to solution- electrospinning.

The scaffolds are characterised and tested for their osteogenic potential in vitro

and in vivo after fabrication following a development process shown in Figure 1.1.

Figure 1.1 General development process of bone TE scaffolds. The process normally starts from fabricating the scaffolds which are rigorously tested in vitro and in vivo step by step before they are clinically relevant. Histology forms the last but critical chain of the assessment.

Generally, in vivo examination, which utilises pre-clinical animal models to

learn the progress of tissue regeneration once a scaffold is implanted in the body

system, is considered to be a true indicator of a TE scaffold’s performance. Histology

is one of the most important post-explant analytical methods in biology and medicine,

and it has been widely applied in bone tissue engineering for bone regeneration

assessment [6]. Histology consists of the microscopic analysis of two-dimensional

tissue sections via a sequential procedure of fixation, dehydration, clearing and

infiltration and embedding (referred here as “processing”); followed by sectioning and

staining [7]. Even though a variety of embedding and sectioning techniques have been

developed in the last two decades, it still remains a challenge to process samples from

bone healing experiments owing to the heterogeneity of the tissues formed during bone

regeneration [8]. Considering the number of animals that can be used in these studies

is also limited, the samples obtained from these in vivo studies are extremely precious.

Thus, it is imperative to select the right analytical techniques to obtain the most

information from limited specimen sections. In this PhD project, I have compared and

optimised the histological techniques for bone specimens embedded in paraffin,

Page 22: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

4 Chapter 1: Introduction

Methyl Methacrylate (MMA) and Technovit 9100 NEW® resin. The outcome of this

study is the standardization of the advanced histological analysis for the future

assessment of bone tissue engineering scaffolds in vivo. Owing to the scaffold

development component of this thesis, it was deemed crucial to also develop advanced

histology techniques to enable the scaffold to be eventually assessed in vivo.

Figure 1.2 Overview of PCL/SrBG scaffolds development in this PhD project. Study 1 and study 2 aimed to produce scaffolds with good bioactivity that are suitable for in vivo implantation. Study 3 was carried out in parallel to provide an optimised method for ex vivo assessment of PCL/SrBG scaffolds.

1.2 PURPOSE OF RESEARCH

The purpose of this PhD project was to develop a PCL/SrBG composite scaffold

using the technique of melt-electrospinning, which may be an alternative treatment

option for healing bone defect compared to autografting. The PCL/SrBG composite

possesses good processibility and bioactivity, and was fabricated into porous scaffolds

via electrospinning. Following fabrication and characterisation, a series of in vitro tests

were carried out on these scaffolds to thoroughly investigate their bioactivity and

osteogenic capacity. I then investigated the possibility to increase the percentage of

SrBG content in the composite and even achieve ordered fibre alignment via a novel

hybrid electrospinning. In vitro tests were performed on the optimised scaffolds for

their bioactivity and osteogenic capacity. The aims of study 1 and study 2 in this study

were to produce PCL/SrBG composite scaffolds with sufficient osteogenic capacity to

Page 23: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 1: Introduction 5

induce bone differentiation when implanted in animal model. In parallel, this research

project also aims to advance the histological techniques for the analysis of complex

explants in our pre-clinical models as an initial aim was to implant these scaffolds as

part of this PhD, hence developing analysis technique in parallel was paramount. In

study 3 of this project, the histological methods for bone assessment were optimised

for the in vivo implanted PCL/SrBG scaffolds (Figure 1.2). The findings of this

research project not only built a basic understanding of the bioactivity and osteogenic

capacity of PCL/SrBG composite scaffold which may be suitable for potential clinical

applications, but also provide a viable tool for evaluating the bone regeneration

capacity of these scaffolds.

1.3 SIGNIFICANCE OF RESEARCH

Current clinical treatment for large bone defects relies on bone grafting:

autografting, allografting and xenografting. However, these techniques show

prominent disadvantages such as donor site morbidity, complications, and infections

and are far from satisfying the patients’ needs in many cases. Other strategies have

been introduced to facilitate bone defect healing such as by the addition of growth

factors. Growth factor treatments are commercially available such as bone

morphogenetic protein-2 (BMP-2) and BMP-7 for bone defect repair. Although BMPs

are strongly osteoinductive and effective [9], they do incur high in-hospital costs for

patients [10]. Even worse, in some cases, they cause undesirable heterotopic

ossification and raise safety concerns [11].

Additive manufacturing, also known as three-dimensional (3D) printing, is

widely employed in TE to produce 3D scaffolds with layer-by-layer deposition

instructed by anatomical and architectural information obtained by medical imaging

techniques (e.g. computed tomography and magnetic resonance imaging) [12]. 3D

printing techniques allow production of 3D structure of complex external shapes with

customized internal microstructures of reproducible porosity and interconnectivity

using biomaterials [13]. These features enable 3D printing to be used by tissue

engineers to fabricate tissue-analogous structures to provide patient-specific implants

for defect healing. Melt-electrospinning produces scaffolds like 3D printing but with

much smaller fibre diameter, which in turn means much higher surface area to volume

ratio. As potential bone graft substitutes, bioactive composite scaffolds may be used

in clinical intervention to circumvent issues associated with bone grafting. In

Page 24: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

6 Chapter 1: Introduction

comparison to growth factors, the use of PCL/SrBG composite scaffolds can be

considered as a more economic and safe option [14]. Furthermore, these composite

scaffolds are made of biocompatible materials and they can be thoroughly sterilised,

therefore, the risk of inflammation and infection is minimised. The fact that the SrBG

binds actively to surrounding soft and hard tissues greatly reduced the chance of

fibrous encapsulation often observed in polymeric implants [15].

Ex vivo assessment of scaffolds using histological approaches is a key process

in the scaffold development. Histology is a traditional technique for the study of the

structure and composition of tissues at the microscopic level and is utilized to analyse

bone morphology in the TE field. Histology uses well-established sequential

procedures to visualise tissue morphology with cellular details. However, it remains

challenging to process bone due to the densely calcified components combined with

the large size of explants. In a parallel and complementary research project, I optimised

and compared common bone histological techniques to provide an optimal tool for

bone regeneration analysis. Our findings can also benefit researchers in the TE field to

pre-plan their histology for optimum results.

On the whole, PCL/SrBG composite scaffolds are promising cost-effective

substitutes to autografting for clinical treatment of bone defects. Combined with

advanced 3D printing, we move toward highly effective and personalised implants.

The findings of this project are expected to propel the production of synthetic bone

graft substitutes that can improve the patients’ quality of life, which is the ultimate

motivation of this PhD project.

1.4 THESIS OUTLINE

The overall aim of this PhD project is to develop a novel PCL/SrBG composite

scaffold via melt-electrospinning as a tissue engineering solution for non-load bearing

defects (such as skull defects), as such we will employ a multi-directional approach,

to both develop a complete scaffold and to develop and standardise end point analytical

assessment techniques (histology). The overall aim is divided into three detailed sub-

aims following the research hypothesis stated in chapter 2.

In chapter 3, I present an extensive literature review covering bone structure and

healing, clinical treatment for bone defects, bone tissue engineering, electrospinning

and biomaterials. This chapter helps to understand the impact of bone fracture situation

Page 25: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 1: Introduction 7

and the recent advancement in the tissue engineering field, which contextualize the

need of our research and how this PhD research can contribute to this research field.

Chapters 4 and 5 describe study outcomes of fabrication, characterization and in

vitro osteogenic capacity evaluation of the electrospun composite scaffolds. Chapter 6

describes the development of optimal histological tools for assessing calcified and

complex tissue engineering scaffold in pre-clinical models. The interrelated studies

reported in these chapters were completed and published or are in preparation for

submission.

Finally, the overall findings in chapter 4, 5 and 6 are collectively concluded and

discussed in chapter 7. The limitations of this PhD and recommended future work are

also discussed in this chapter.

Page 26: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 27: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 2: Research Hypothesis and Aims 9

Chapter 2: Research Hypothesis and Aims

2.1 HYPOTHESIS

We hypothesize that the bioactivity of PCL can be increased by incorporating

SrBG particles, and that these PCL/SrBG composites can be produced by melt-

electrospinning into porous scaffolds which are osteoinductive, osteoconductive,

capable of osteointegration and bioresorbable in a controlled rate.

2.2 RESEARCH AIMS

The overall aim of this PhD project was to develop melt-electrospinning as a

tissue engineering solution for non-load bearing defects, as such I employed a multi-

directional approach, to both develop the PCL/SrBG composite scaffold and

standardise end point histological analytical assessment criterion. This overall aim was

broken down to 3 specific aims that formed the basis for three interrelated studies

presented below.

(Study 1) To design, fabricate and characterise PCL/SrBG (10 wt%)

composite scaffolds for bone regeneration via melt-electrospinning, followed by

assessing the in vitro bone regeneration capacity of the composite scaffolds.

In study 1, I attempted to melt-electrospin porous scaffolds of PCL/SrBG

composite which combined the processibility of PCL and the osteogenic properties of

SrBG by mixing SrBG particles into PCL bulk. SrBG particles were ground to <38 μm

with mortar and then mixed into molten PCL by manual stirring until even distributed.

PCL/SrBG composites containing only 10 wt% of SrBG were fabricated into scaffolds

via melt-electrospinning due to the limitations posed by SrBG particle size and

composite mixing technique. These electrospun composite scaffolds were then

characterised and comprehensively assessed for their in vitro osteogenic properties.

Sections 1a – 1c describes the methodology undertaken in study 1.

1a: To optimise melt-electrospinning technique for the fabrication of PCL/BG

composite scaffolds and characterisation of these scaffolds

10 wt% PCL/SrBG composite was manually prepared by stirring the mixture of

SrBG particles and PCL bulk at 65 °C on a hot plate. The melt-electrospinning

Page 28: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

10 Chapter 2: Research Hypothesis and Aims

parameters (i.e. temperature, voltage, polymer flow rate and collector to spinneret

distance) for the fabrication of PCL and PCL/SrBG composite scaffolds were

optimised for the built-in-house melt-electrospinning rig. The fabricated PCL/SrBG

composite scaffolds and control PCL scaffolds were characterised with micro-

computed tomography, light microscopy and scanning electron microscopy (SEM) for

their properties such as fibre diameter, surface topography and SrBG particle

distribution.

1b: In vitro bioactivity evaluation and comparison of both PCL and PCL/SrBG

scaffolds

Two mechanisms account for the bioactivity of SrBG: tissue bonding via the

formation of a hydroxycarbonate apatite (HCA) layer & ion dissolution of the SrBG

[16]. To observe the HCA layer formation and measure the ion concentration, the

scaffolds were immersed in serum-free culture media for up to 28 days incubated at

37 °C. At predetermined time points, the scaffolds were retrieved from the media for

SEM and energy-dispersive X-ray spectroscopy (EDX) analysis, while the remaining

media were analysed with Vista MPX Inductively Coupled Plasma - Optical Emission

Spectrometer (ICP-OES) to quantify the concentration of Ca2+, PO43-, Si4+ and Sr2+.

1c: Investigation of the in vitro osteogenic capacity of PCL and PCL/SrBG

scaffolds.

MC3T3-E1 cells, a mouse osteoblast precursor cell line, were seeded onto both

PCL and PCL/SrBG scaffolds and cultured in normal growth media and osteogenic

media (media supplemented with osteogenic factors). At predetermined time points,

the scaffolds with cells were retrieved and assessed with a series of qualitative and

quantitative assays. The cell morphology and attachment on scaffolds were assessed

with confocal laser scanning microscopy (CLSM) and SEM. The scaffolds

cytotoxicity were assessed with LIVE/DEAD and MTT (3-(4,5-Dimethylthiazol-2-

yl)-2,5-diphenyltetrazolium bromide) assays. Cell proliferation and osteoblastic

differentiation were quantified by PicoGreen assay normalised alkaline phosphatase

(ALP) assay and polymerase chain reaction (PCR) assay. In addition, the scaffolds

with cells were stained with Alizarin red S and Van Gieson' stain for the assessment

of matrix mineralisation and matrix formation, respectively.

Study 1 has been completed and published:

Page 29: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 2: Research Hypothesis and Aims 11

Ren, Jiongyu et al. 2014. “Melt-Electrospun Polycaprolactone Strontium-

Substituted Bioactive Glass Scaffolds for Bone Regeneration.” Journal of Biomedical

Materials Research - Part A 102(9):3140–53.

This study is presented in details in chapter 4.

(Study 2) To optimise the composite scaffold design and to further increase

SrBG filler percentage in PCL to enable greater osteoinductivity.

Here I aimed to improve bioactivity of PCL/SrBG composite by increasing the

percentage of SrBG in PCL. In study 1, we found that PCL/SrBG composite scaffolds

exhibited in vitro bioactivity and enhanced osteogenic capacity. However, these

composite scaffolds were not yet ideally osteogenic due to an insufficient amount of

SrBG in PCL (only 10% by weight). Also, the scaffolds were melt-electrospun into

circular sheets with random fibre layout. In order to improve the control of fibre

deposition and improve SrBG yield in the PCL/SrBG composite, the hardware design

of a new electrospinning rig was significantly upgraded including a motorised stage

and dual high voltage power packs, and the size of SrBG was reduced from ≤ 38 μm

to ≤ 6 μm (details reported in Chapter 5).

Our theory that increased SrBG in the composite may lead to enhanced

osteogenic capacity is based on the fact that polymer/BG composite materials

exhibited higher bioactivity when higher amount of BG presented in them, as indicated

by more rapid in vitro calcium phosphate layer formation [17].

2a: To optimise the preparation technique of PCL/BG composites with higher

weight percentage of SrBG filler phase

The SrBG particles were ground from 100 μm down to < 6 μm prior to composite

preparation with a micronizing mill to reduce the risk of needle blockage. In addition,

the reduced particle size also increased the total surface area of the SrBG particles.

The PCL/SrBG composites were prepared by incorporating fine SrBG particles into

the PCL bulk using solvent precipitation technique instead of manual stirring in order

to ensure even distribution of SrBG particles within PCL bulk. The SrBG filler phase

was incorporated into PCL bulk as high as 50 wt% for enhanced bioactivity of the

PCL/SrBG composite.

Page 30: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

12 Chapter 2: Research Hypothesis and Aims

2b: To fabricate 50 wt% PCL/SrBG with controlled fibre alignment via a novel

hybrid melt-electrospinning technique in a direct writing mode, followed by scaffold

characterization

The 50 wt% PCL/SrBG composite scaffolds were fabricated using a novel

solvent assisted hybrid melt-electrospinning technique. This hybrid electrospinning

technique utilized solvent to reduce the viscosity of composite melt to facilitate

establishing a continuous and stable polymer jet that were deposited on the collector

in a 0/90 º cross hatch laydown pattern.

Light microscopy and scanning electron microscopy (SEM) were used to

characterise surface topography and fibre diameter. Mechanical stiffness of PCL/SrBG

fibres was assessed with atomic force microscopy. Inductively coupled plasma mass

spectrometry (ICP-MS) technique was used to analyse the elemental concentration of

specific ions in the dissolution cell culture of the composite scaffolds.

2c: Investigation of the in vitro osteogenic capacity of PCL and PCL/SrBG (50

wt%) scaffolds.

MC3T3-E1 cells were used again to study the effect of PCL/SrBG (50 wt%)

scaffolds when they were in contact with cells, the PCL scaffolds were studied as

control. The cells seeded on both type of scaffolds were cultured in normal growth

media and osteogenic media (media supplemented with osteogenic factors). Similar in

vitro assays with study 1 were carried out to examine the bioactivity and osteogenic

capacity of the PCL/SrBG (50 wt%) scaffolds and to make comparison to PCL/SrBG

(10 wt%) scaffolds produced in study 1. At predetermined time points, the scaffolds

with cells were retrieved and assessed with a series of qualitative and quantitative

assays. The cell morphology and attachment on scaffolds were assessed with CLSM

and SEM. The scaffolds cytotoxicity were assessed with LIVE/DEAD and MTT

assays. Cell proliferation and osteoblastic differentiation were quantified by

PicoGreen assay normalised ALP assay and PCR assay.

(Study 3) To optimise and standardize histology techniques for assessing

bone/TE bone

As a scaffold for bone, the composite implant is expected to contain both

mineralised and soft tissues in vivo, creating differentially dense interfaces. The

complex calcified tissues coupled with often large explant sizes make it challenging

Page 31: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 2: Research Hypothesis and Aims 13

for their histological assessment. In order to advance our pre-clinical models and post

explant analysis of PCL/SrBG scaffolds, we optimised and compared two most

common bone histological techniques: paraffin embedding (after decalcification) and

resin embedding in either methyl methacrylate (MMA) or Technovit 9100 NEW® in

study 3 in parallel scaffold fabrication. Four commonly used histological stains for

bone were investigated including Haematoxylin & Eosin (H&E), Goldner’s trichrome

staining, Von Kossa staining and immunohistochemistry (IHC). To draw a

comprehensive conclusion, I investigated tissues and implants from commonly used

preclinical models in TE: sheep, pig, rat and mouse.

3a: Optimisation of commonly used histological stains for resin embedded

specimens.

The bone specimens embedded in MMA and Technovit resin were sectioned via

Exakt cutting and grinding system, referred as ground sectioning, producing sections

of around 27 μm. Despite the above-mentioned stains were traditionally used, the

staining protocols were not readily applicable to the ground sections mainly due to the

hydrophobicity of resin. Therefore, firstly I optimised the staining techniques for resin

ground sections. The optimised staining protocols are presented in detail in chapter 6

which other researchers can easily follow.

3b: Comparison of H&E, Goldner’s trichrome staining, Von Kossa staining

and IHC staining techniques for the assessment of mineralised tissues originated

from sheep, pig, mouse and rat processed via paraffin and resin embedding and

sectioning routes

The ground sections of resin embedded specimens (MMA and Technovit) and

microtome sectioned paraffin embedded specimens were stained with H&E, Goldner’s

trichrome staining, Von Kossa staining and IHC stains, and compared accross all 4

species of sheep, pig, mouse and rat. Overall resin ground sectioning provided the

highest quality outcome in both complex (mouse paw) and large sections (sheep tibia).

Two disadvantages of ground sectioning are the relatively large specimen consumption

per section and long processing time. Paraffin sections showed good bone morphology

but poor hard/soft tissue interface preservation and no bone mineral information.

Goldner’s Trichrome staining on ground resin sections provides the best

differentiation between hard and soft tissue with clear cellular details. IHC worked

with paraffin microtome sections and ground sections of Technovit embedded

Page 32: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

14 Chapter 2: Research Hypothesis and Aims

specimens. These results can guide researchers to preplan their histology for optimum

results.

Page 33: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 15

Chapter 3: Literature review

3.1 BONE BIOLOGY

3.1.1 Structure of bone

Bone is a highly specialized organic/inorganic composite fabricated by nature.

As a complex connective tissue, bone consists of three components: the cellular

components, the hydrated extracellular organic matrix (mainly collagen,

approximately 35% dry weight), and the extracellular mineral phase (mainly

hydroxyapatite, 65% dry weight) [6,18,19]. The major component of the bone organic

extracellular matrix (ECM) is type I collagen, this accounts for approximately 90% of

the total volume of the ECM, with the rest formed from other minor collagens such as

type III and type V collagen and a variety of non-collagenous proteins, glycoproteins

and proteoglycans in relatively small quantities [6,20]. The molecular structure of

collagen is a stabilized left-hand superhelix fibril, and bundles of fibrils comprise

nano-size collagen fibres that provide bone with great tensile strength (a Young’s

modulus of 1-2 GPa and ultimate tensile strength of 50-1000 MPa) [6,18]. The

inorganic phase of bones’ ECM further enhances the stiffness and compressive

strength of the bone. The principle component of this organic phase is a calcium

phosphate based crystalline structure called hydroxyapatite (HA) with the chemical

formula of [Ca10(PO4)6(OH)2] (a Young’s modulus of 130 GPa and an ultimate tensile

strength of 100 MPa) [6,18]. The bonding between the organic matrix and inorganic

hydroxyapatite (HA) crystals makes bone a natural bio-composite material with

excellent mechanical properties. Weiner & Wagner provided an excellent overview of

bone linking its macro-structure and micro-structure by breaking down the bone

structure to seven levels of hierarchy (Figure 3.1) [18]. Nanoscopically, HA platelets

orient and align within self-assembled collagen fibrils, making the first level of bone

hierarchical structure. The mineralised collagen fibrils are the basic building block of

bone. They stack parallel as lamellae which are then arranged concentrically around a

central Haversian canal to form the basic unit of bone, Osteon. Finally, the macro-

structure of bone is formed by osteons: either densely pack into compact bone, or form

a trabecular network into cancellous bone [18,20].

Page 34: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

16 Chapter 3: Literature review

Figure 3.1 The seven hierarchy levels of bone structure, as demonstrated by Weiner and Wagner [18]. Level 1 shows the two major components of bone: mineral particles (left) and collagen fibril (right). Level 2 shows a mineralized single collagen fibril which combines the two phases. These mineralized fibrils organize into arrays as shown in level 3 and level 4 and

Page 35: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 17

eventually forms the basic unit of bone – osteon (level 5). Level 6 shows the two types of bones: the compact cortical bone and the porous spongy bone. Level 7 is an overview of a whole long bone.

Generally, bones are classified by their shape into several distinct types: long,

short, flat, irregular and sesamoid [21]. The typical structure of a long bone is shown

in Figure 3.2. When looking into their macroscopic structure, bones are not uniform,

but are composed of a hard outer layer, compact bone (or called cortical bone) with an

average density of 0.2 g/cm3 and an interior more porous part, trabecular bone (also

called cancellous or spongy bone) with an average density of 1.80 g/cm3 [20,22]. As

shown in Figure 3.2, both the inner and outer surfaces of the bone are covered by thin

vascular membrane-like layers: the outer membrane is named periosteum and the inner

membrane is called endosteum [23]. Both the periosteum and endosteum play essential

roles in bone growth and repair by providing a continuous supply of osteoprogenitor

cells or new osteoblasts [21].

Figure 3.2 Schematic illustration of long bone and its micro-structure [23]

The Haversian canal is essential for bone function as it contains nerve and blood

vessels. The blood vessels are responsible for nutrient supply and waste disposal for

the cells residing in the bone matrix [6,23]. Four types of cells populate the mineralised

bone matrix: osteoblasts, osteocytes, osteoclasts and bone lining cells [23]. Derived

from local osteoprogenitor cells, fully differentiated osteoblasts are responsible for

bone matrix regulation and membrane-associated alkaline phosphatase (ALP)

synthesis. Osteoblasts are considered differentiated into osteocytes once they are

surrounded by mineralized matrix or lacunae [6]. Due to the pivotal role of osteoblasts

Page 36: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

18 Chapter 3: Literature review

in bone formation, the clinical success of implanted polymers in bone defects largely

depends on the attachment of osteoblasts to the material surface. Any biomaterial

which is to be used for the repair of a bone defect must be conductive to osteoblast cell

attachment, proliferation and differentiation in order for bone regeneration to occur.

Osteoclasts, on the contrary, are responsible for mineralized bone matrix resorption by

secreting acids and enzymes to degrade both organic and inorganic constituents of the

bone [23]. The synergistic actions of osteoblasts and osteoclasts are the basis of a

dynamic bone remodelling process whereby bone is constantly remodelled in order to

maintain its unique architecture throughout the life [6].

3.1.2 Bone fracture healing

Unlike the fibrous scar of the soft-tissue healing, bone has a remarkable self-

healing capacity characterised by its unique scarless repair. The repair process includes

four overlapping stages as shown in Figure 3.3 [24]. Briefly, the hematoma formation

stage (Figure 3.3a) is the first post-fracture stage which keeps the fracture area in an

enclosed environment. The hematoma is formed around the broken bone ends by blood

from adjacent structures due to disrupted blood vessels. Angiogenesis (formation of

new blood vessels) takes place when several types of cells are recruited to the defect

site to eventually form fibrocartilage at this stage. Simultaneously the periosteum

enables direct bone formation and an external callus is created as a result (Figure 3.3b);

Subsequently, a hard bony callus is formed from within through the mineralisation of

the internal callus (Figure 3.3c); Finally, the remodelling activity of the bone replaces

the hard callus with lamellar bone and also reduces the callus size at the fracture site

[24]. After these repair stages, the original biochemical and biomechanical properties

of the bone are fully restored as well as the vascular supply. It is reported that the

regenerated bone shows no difference to undamaged host bone tissue [25].

Page 37: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 19

Figure 3.3 The four stages of bone fracture regeneration [24]

3.2 CURRENT TREATMENTS FOR LARGE BONE DEFECTS

Despite the exceptional regenerative capacity of bone, the healing of large

segmental defects that exceeds the body’s self-healing capacity (e.g. a gap over 2.5

times the bone radius in long bone defects [26]), such as those caused by congenital

disorder, traumatic injury and tumor removal, does not take place without surgical

intervention [27]. Bone reconstruction still remains a challenge in these conditions

[27]. This situation is referred to as a critical-size defect [23,27]. In critical-size bone

defects, it is necessary to implant grafting materials in order to bridge the gap. To

achieve positive regenerative results, the implanted material should ideally possess the

following properties: an osteoconductive structure, osteoinductivity, biocompatibility

and biodegradability. Favourably, the implant should provide structural support and

stability [9]. In the clinical practice, the treatments for large bone loss rely on bone

grafting, including autogenic (bone graft from patients own body), allogenic (bone

graft from a donor body) and xenogenic (bone graft from a different species to human)

bone grafting, to restore bone loss due to trauma or above mentioned conditions [9,27].

Every year, around 4 million bone grafting or bone substitutes surgeries are performed

worldwide, which again highlights the demand in the surgical demand as well as the

impact of critical-size bone defects [27].

3.2.1 Bone grafting

Among the bone grafting techniques, autografting, which is to graft bone tissue

from one site to another within an individual, is considered the ‘gold standard’ for

critical-size bone defect reconstruction [27,28]. Autografts are ideal for bone

reconstruction as they are not only osteoconductive but also osteoinductive [28]. It is

Page 38: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

20 Chapter 3: Literature review

reported that the use of autografting has been successful in treating large bone defects

with no risk of disease transmission from a donor tissue (whereas allografting may

pose disease transmission problems) [27]. However, the disadvantages of autografting

are also prominent. Firstly, because the amount of donor bone is limited, it is often

difficult or sometimes impossible to harvest enough bone to graft in the situations of

large or multiple bone defects. Secondly, the harvesting procedure normally leads to

donor site morbidity and also a great pain for the patient. Thirdly, complications often

exist after the surgery such as infection, hematoma and in some cases pelvic instability

[28]. To overcome these difficulties, allograft (to transplant bone tissue from cadavers

or living donors) and xenografts (to transplant bone tissue from other species) have

been introduced to the clinical practice as alternatives to autograft. Allograft bone is

osteoconductive, it supports bone formation, and most importantly, the supply is much

less limited compared to autografting. Although it is possible to achieve large bone

defect structural restorations, the application in the clinical practice is limited due to

its poor osteogenic and osteoinductive capacity, large immunogenic response, limited

revascularization and the potential for disease transmission [29]. Additionally, results

revealed that newly formed bone with allograft procedure is not likely to incorporate

into the host [30]. Xenogenic graft is rarely used because of the high failure rate [29].

3.3 BONE TISSUE ENGINEERING

Since bone grafting is not able to consistently provide satisfactory results,

researchers have endeavoured to seek more effective alternatives. One avenue of

research has led to the emergence of the field of Tissue Engineering (TE). The term of

‘TE’ was first created in the mid-1980s but was not well established until late 1980’s

by a prominent research group in the US, who gave the first definition of tissue

engineering [31]. The definition of ‘TE’ developed with the evolution of the research

field, and was described by Robert Langer and Joseph P. Vacanti in their review paper

published in Science on May 14, 1993 [32]. According to their definition, tissue

engineering is ‘an interdisciplinary field that applies the principles of engineering and

the life sciences toward the development of biological substitutes that restore,

maintain, or improve tissue function’ [32]. Over the years, the research field of TE has

grown tremendously, especially after the development of three-dimensional polymeric

scaffolds for tissue or organ regeneration [33]. Not only does the field of TE hold great

promise for the future of medicine, but it has become a billion dollar industry and has

Page 39: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 21

created almost 14,000 employment opportunities worldwide [34]. Additionally, the TE

industry shows an increase in growth lately as more and more TE products are being

trialled and applied to clinical practice, with a number of them already on the market

(e.g. INFUSE® Bone Graft (Medtronic) which is a bone graft material used in

treatments of certain types of spinal surgery, acute tibia fracture and oral maxillofacial

defects).

3.3.1 3D printing scaffolds

Since the first technique of producing solid three-dimensional (3D) structure was

introduced in the 1980s [13], a series of 3D printing techniques were introduced and

these techniques have been applied in a diversity of fields including TE [35,36].

Typical 3D printing, e.g. fused deposition modelling (FDM) [37], stereolithography

(SLA) [38], selective laser sintering (SLS) [39], selective laser melting (SLM), direct

metal deposition (DMD) and inkjet printing, can produce many designs of complex

geometries in a layer-by-layer deposition based on specific 3D models [35]. 3D

printing scaffolds have the advantages of accurately and precisely controlled pore size

and geometry over scaffolds manufactured with traditional techniques [37,38,40,41].

The features of 3D printing make them potential custom-made solutions for patients

by providing scaffolds that precisely match the defects using medical scanning data

injury site.

3.3.2 Tissue engineering scaffolds

As stated in the introduction, millions of people are affected by bone defect-

related diseases and there is a pressing need for better bone substitutes worldwide [42].

The bone TE field seeks to address the need by introducing engineered implants which

will eventually become integrated within the patients, thus providing potentially

permanent cures. Tissue engineered constructs, often termed as ‘scaffolds’, are 3D

structures that serve as temporary templates to guide cell attachment, proliferation,

differentiation and extracellular matrix (ECM) formation at defect sites. A typical

workflow for the proposed scaffold-based approaches to bone tissue engineering

involves 5 basic steps: i) 3D data acquisition through medical imaging techniques at

the time of diagnosis, such as computed tomography (CT) and magnetic resonance

imaging (MRI); ii) 3D model generation of the defect anatomy on computer by

converting the medical scan data with software such as computer-aided design (CAD);

iii) the 3D image of the final defect anatomy is then translated into machine language

Page 40: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

22 Chapter 3: Literature review

to manufacture a biodegradable scaffold, usually made from a resorbable polymer; iv)

then seed the scaffold with cells and/or possibly supplement with bioactive molecules

(e.g., Bone morphogenetic proteins (BMPs)); v) finally the scaffold is implanted into

the defect site to induce and direct the growth of new bone [12,23]. The whole process

is illustrated in figure 3.4. Ideally, the cells will attach onto the scaffold, proliferate,

differentiate and produce bone matrix as the scaffold degrades, eventually, heal the

bone defect.

Figure 3.4 Schematic overview of the scaffold-based approach to bone TE [12]

3.3.3 TE scaffolds specifications

Fundamentally, bone TE promotes the body’s natural regenerative capacity in

defect repair with engineering principles, which is termed bone biomimetics [23]. For

this purpose, ideal synthetic bone scaffolds must address the following biomimetic

requirements. i) provide temporary mechanical support to the defect site and act as

substrate for bone deposition; ii) contain interconnective pores within the architecture

to allow for rapid vascularisation and bone growth and to prevent unwanted tissue

growth, mainly soft tissues, into to the wound bed; iii) be osteoconductive and

Page 41: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 23

osteoinductive; iv) promote integration between host bone and regenerated bone; v)

be bioresorbable without toxic degradation products and degrade in a controlled

manner; vi) be capable of retaining bioactivity after sterilization and free of

inflammatory response; vii) potentially have controlled drug delivery capacity

[23,37,43]. To meet these requirements, it is critical for tissue engineers to select the

right material and proper processing technique for scaffold fabrication.

3.4 SCAFFOLD MATERIALS

The composition of scaffold materials plays a critical role in the overall success

of bone TE treatment because it is essential to mimic the biochemical and biophysical

properties of the native tissue. The ideal material must be non-toxic, biocompatible,

biodegradable, and must possess appropriate mechanical properties for load bearing

applications. Additionally, they should ideally be osteoinductive and osteoconductive

[23,43]. The development of synthetic bone scaffold materials has increased over the

years with a variety of candidates investigated, including metals, ceramics, polymers

and their composites. Due to their superior mechanical and osteogenic properties over

other materials, a paradigm shift has taken place towards the use of polymers,

bioceramics and their composites as the principle materials for bone scaffold

manufacturing [44].

3.4.1 Polymers

To date, both natural polymers and synthetic polymers have been investigated as

bone scaffold candidates. Although both types of polymers have been investigated for

TE, synthetic polymers are being more widely used because of their tunable

degradability, higher purity levels and better mechanical properties than natural

polymers [23]. Common synthetic polymers for bone repair include polyesters,

polydioxanone, poly (ethylene glycol) (PEG), poly (propylene fumarate) (PPF), poly

(orthoesters), polyanhydrides, and polyurethanes [23]. The properties of some

example polymers are summarized in Table 3.1.

Page 42: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

24 Chapter 3: Literature review

Table 3.1 Properties of biodegradable polymers as bone scaffold materials [35,36] Polymer Thermal and Mechanical Properties Approx.

Degrade time (months)

Areas of application Melting

point (°C)

Glass Transition (°C)

Approx. Strength

Poly (lactide) 173-178 60-65 2.7GPa (Modulus)

6 to12 Orthopaedic Surgery Oral and Maxillofacial surgery

Poly (glycolide) 225-230 35-40 7.0GPa (Modulus)

>24 Orthopaedic surgery, General surgery, Sutures

Polydioxanone 58-63 -65-60 >24 Orthopaedic surgery General surgery Sutures

Poly (D,L-lactide-co-glycolide) Poly (D,L-lactide-co-glycolide) 85/15 Poly (D,L-lactide-co-glycolide) 82/18 Poly (D,L-lactide-co-glycolide) 1090

Amorphous 50-55 2.0GPa (Modulus)

5 to 6 Suture Drug Delivery Oral and Maxillofacial surgery, General surgery, Sutures, Drug delivery, Periodontal surgery

Polycaprolactone 58-63 -65-60 0.4 GPa (Modulus)

>24 Drug delivery, Sutures

Polycaprolactone (PCL) is a semi-crystalline polyester that belongs to the

family of aliphatic polyesters [45]. As shown in Table 3.1, PCL has a glass transition

and melting temperature of approximately -60 °C and 60 °C respectively [23,45]. It

stands out from other biodegradable polymers because of its exceptional biochemical

and physio-chemical properties [45]. From the Bone TE perspective, PCL has the

following advantages: (i) a low melting point meaning PCL has superior rheological

and viscoelastic properties which make the polymer easy to shape and manufacture

into a variety of scaffold shapes; (ii) PCL is biocompatible and non-toxic and has been

approved in a number of biomedical devices by the US Food and Drug Administration

(FDA); (iii) not only is PCL biodegradable, but it is also bioresorbable which means it

Page 43: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 25

may resorb over time and be excreted through a citric acid cycle, resulting in total

elimination of PCL with no residual side effects (process shown in Figure 3.5) and (iv)

good solubility and blend-compatibility with other polymers such as cellulose

propionate, cellulose acetate butyrate, polylactic acid and polylactic acid-co-glycolic

acid [45].

Although PCL has the potential to be a strong candidate for a bone TE solution,

its application is limited mainly because of its hydrophobicity and lack of bioactivity

[33,46]. However, PCL scaffolds can be made more hydrophilic with sodium

hydroxide (NaOH) etching [47]. Like a typical polymer, PCL is considered ‘soft’ for

high load bearing applications because of its deficiency in the compressive modulus

[23]. Additionally, the long-term degradation (up to 3-4 years for homo-PCL to

completely resorb) and further prevent PCL from widespread applications in bone TE

[48]. Lastly, PCL is osteoconductive but not considered osteoinductive. In order to

overcome these shortcomings of PCL, we are seeking materials that can be

incorporated into the polymer bulk to render them more suitable. One approach is to

incorporate a high modulus micro and/or nano-scale bioactive material (e.g. bioactive

glass) to introduce bioactivity to PCL bulk and enhance its mechanical properties.

Figure 3.5 The degradation and elimination pathway of PCL. PCL formed hydrolytic intermediates 6-hydroxylcaproic acid and acetyl coenzyme A, which is eliminated from the body via the citric acid cycle [45]

3.4.2 Bioactive glass (BG)

BG is a subgroup of inorganic biomaterials based on silicate or phosphate

systems, which exhibit unique osteoinductive properties and show good bonding to

both bone and soft tissues in biological fluid [16,49]. Since Hench and his colleagues

developed the first bioactive glass in 1969, known as Bioglass® or 45S5 [48,50,51], a

Page 44: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

26 Chapter 3: Literature review

variety of different bioactive glasses (BGs) have been developed and applied in bone

TE [16,52]. Bone bonding and ionic dissolution are the two mechanisms that account

for the bioactivity of BGs. The BGs bond strongly to bone tissue through a precipitated

layer of hydroxycarbonate apatite (HCA), which interacts with collagen matrix of bone

[53].

Mechanism of HCA layer formation

The formation of HCA is well understood, and Larry L. Hench revealed this

process of HCA precipitation on 45S5 BG (with a composition of 46.13 SiO2–2.60

P2O5–24.35 Na2O–26.91 CaO (mol%)) in 5 main stages shown in Table 3.2 [16,51].

Table 3.2 Reaction stages of HCA layer formation on 45S5 BG [54] Stages Reaction

1 Rapid ion exchange of Na+ or Ca2+ in BG with H+ from solution: Si – O – Na+ + H+ + OH- Si – OH+ + Na+ + OH-

2 Stage 1 leads to loss of silica in the form of Si(OH)4 and increase of pH and breaking of Si-O-Si bonds and form Si-OH (silanol bonds): Si-O-Si + H2O Si-OH + OH-Si

3 Si-OH groups condensate and repolymerise to SiO2-rich layer:

4 Migration of Ca2+ and PO43- groups to the surface through the SiO2-rich layer leads

to the formation of an amorphous CaO-P2O5 layer, followed by the growth of CaO-P2O5-rich film by incorporation of soluble calcium and phosphates from solution.

5 Crystallization of the amorphous CaO-P2O5 film by incorporation of OH-, CO32-,

or F- anions from solution to form a mixed hydroxyl, carbonate, fluorapatite layer.

Ionic dissolution

Once the HCA layer has formed, it absorbs proteins and then enhances cell

attachment, differentiation and bone matrix production [16]. Ionic dissolution products

through HCA are believed to be a key element in the osteogenesis of BGs, however,

the detailed mechanism of how BGs enhance osteogenic responses is still largely

unknown. Figure 3.6 shows the typical ions released from BGs and the biological

responses to these ions [49].

Page 45: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 27

Figure 3.6 The effect of bioactive glass ion dissolution products on biological responses [49]

Strontium-substituted bioactive glass

Strontium ions have been found to be beneficial to patients suffering from

osteoporosis. Sr-doped products are already available on the market, for example, oral

administration of Sr-ranelate, under the trade name Protelos™, has been widely used

in osteoporotic patients and received satisfactory clinical outcomes [55,56]. In 2010,

Professor Molly Stevens’s research group developed a class of strontium-substituted

bioactive glasses (SrBG) by replacing 0-100% of the calcium components with

strontium in traditional 45S5 BG [57]. The resulting SrBGs showed enhanced

bioactivity compared to 45S5 BG [57,58]. As reported, a critical concentration of Sr

ions in the dissolution medium plays a significant role in enhancing bone cell activity

[49,57–59]. In their studies, SrBG not only promoted osteoblasts proliferation and

alkaline phosphatase (ALP) activity when directly applied to human osteosarcoma cell

line, Saos-2, but also inhibited osteoclasts differentiation and decreased tartrate

resistant acid phosphatase (TRAP) activity. This implicates that the SrBG not only

enhance new bone formation but also suppress bone resorption by osteoclasts, leading

to increased turnover of new bone regeneration. In a recent in vitro study using

mesenchymal stromal cells, SrBG was found to upregulate osteogenic genes starting

with BMP-2 which then led to a cascade of gene up-regulation including Runt-related

Page 46: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

28 Chapter 3: Literature review

transcription factor 2 (Runx2), alkaline phosphatase (ALP), collagen type I (ColI) and

osteocalcin [60]. The molecular mechanisms regarding how the incorporation of Sr2+

promoted osteogenic differentiation were proposed as the activation of two cell

signalling pathways: the Ras_MAPK and Wnt-Catenin [60–62]. The advantages of

SrBG have made them potential candidates for a wide range of applications in

orthopaedic regenerative medicine. However, the innate brittle nature of BG makes it

very difficult to process it into useful 3D scaffold with adequate mechanical strength

and porosity [48], thus the application of BGs alone is limited in bone TE.

When looking at both PCL and SrBG, we can see that they have complementary

properties. PCL can be fabricated into 3D scaffolds with desired structure and adequate

mechanical strength, but it lacks bioactivity; while SrBG has great osteogenic capacity

but brittle in nature. Therefore, by incorporating SrBG micro/nano- particles into

PCL we believe that we can generate a superior composite material in which the

inorganic phase will act to improve the mechanical strength and the biological

properties of polymeric fibers, such as cell compatibility, osteogenic differentiation,

and bone matrix calcification [63]. Such composite materials will be detailed in the

following section in the context of bone regeneration.

3.4.3 Biodegradable polymer/bioactive glass composites

Facing a complex system like human bone, conventional single-component materials

are not capable of satisfying all the criteria for a bone replacement/regeneration

scaffold. To fulfil as many requirements as possible, composite systems of

biodegradable polymers and ceramics are being developed, and hold promise as a bone

TE scaffolds. In these composite systems, micro- or nano-scale ceramic particles are

incorporated into biodegradable polymers to take advantage of their processibility

[16]. The addition of an inorganic phase to bioresorbable polymers not only

counteracts their poor bioactivity, but also accelerates the polymer degradation rate by

increasing the hydrophilicity and water absorption of the normally hydrophobic

polymer matrix [16,50]. Furthermore, in addition to improving mechanical properties,

the cations released from BG filler phase buffers the local pH which may have been

affected by the acidic polymer degradation products [16]. Coating BG particles onto

polymer scaffold surface [64] or coating polymer onto BG scaffold surfaces [65] were

alternative methods to produce composite scaffolds. Because of the associated

disadvantages of the coating techniques such as clogging of pores with the scaffolds

Page 47: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 29

and weak bonding between the organic and inorganic phases [50,66], this review

would not introduce these techniques extensively. To our knowledge, the number of

studies on polymer/bioactive glass composites as bone scaffolds is relatively limited.

In vitro studies carried out by Boccaccini et al. in 2003 confirmed that by filling their

poly(DL-lactic acid) foam scaffolds with BGs the osteogenic activity and

mineralization of human osteosarcoma cells (MG-63) were greatly promoted [67,68].

This result was in accordance with earlier studies conducted by Lu et al. [68], in which

porous composite scaffolds of polylactide-co-glycolide (PLAGA) and 45S5 bioactive

glass showed higher levels of Type I collagen synthesis and mineralization of human

osteosarcoma cells (SaOs-2). While most of the scaffolds used in these studies were

manufactured with traditional TE techniques (summaries in Table 3.3), the use of

additive manufacturing methods to produce 3D scaffolds has recently gained

momentum in the TE field.

Nano-sized BG particles or nanofibres have been reported to exhibit advantages over

their micro-scale counterparts. Compared with micro-sized particles of the same total

volume, BG nanoparticles have much larger surface area relative to their volume

which could contribute to improved bioactivity compared with micro-sized particles.

This was confirmed by S.K. Misra et al. [69] who investigated the in vitro activity,

protein adsorption capability and mechanical properties of both micro- and nano-sized

BG/poly (3hydroxybutyrate) composites (10 – 30 wt%). The results of these studies

indicated that the incorporation of nano-sized BGs not only reinforced the mechanical

stability of the composites, but also induced nanotopography on the scaffold surface

hence improved total protein adsorption and bioactivity of the composite [69–73].

Fabbri et al. reported that the mechanical properties of PCL/45S5 BG scaffolds (with

0 - 50 wt%), fabricated by a solid-liquid phase separation method, were enhanced with

higher amount of the BG loading in the scaffolds [74]. Further in vivo data were

obtained by Jo et al., which revealed good bone-forming ability of PCL/nano-BG

composite in a rat calvarial bone defect model [75]. Rowe et al. produced PCL/borate

bioactive glass membranes with up to 10 wt% BG loading using solution-

electrospinning technique [76]. These composite electrospun membranes were found

to support osteoblast attachment and proliferation.

Page 48: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

30 Chapter 3: Literature review

PCL/SrBG composite scaffolds

Because of the favourable properties of PCL and SrBG as bone implant materials, the

PCL/SrBG composite material has been made into scaffolds and explored for their

bioactivity by other researchers [14,77]. The composite material of PCL/45S5 BG and

PCL/SrBG were produced into 3D scaffolds with BG loading of 10% [14] and 50%

[77] using the melt extrusion based additive manufacturing techniques, resulting in

scaffolds with fibre diameters of ~ 500 μm (estimated based on the SEM images). The

PCL/SrBG scaffolds with 10% SrBG loading showed in vitro bioactivity and had

higher compressive strength compared to PCL scaffolds. However, in vitro testing of

these scaffolds showed the PCL/SrBG (10%) scaffolds were not able to stimulate

osteoblast differentiation in the absence of osteogenic cytokines (typically ascorbic

acid, β-glycerophosphate, and dexamethasone).

In order to further enhance the bioactivity of the composite scaffolds, Poh et al. then

increased the SrBG loading to produce PCL/SrBG scaffolds with 50wt% SrBG loading

[77]. In vitro testing was performed on these scaffolds and the PCL/SrBG scaffolds

were found to be able to stimulate osteoblast differentiation even in the absence of

osteogenic supplement in the cell culture media. The osteogenic properties of the

PCL/SrBG scaffolds were indicated by the up-regulation of osteopontin (OPN) and

osteonectin (OCN), two genes expressed at different stages of osteoblast

differentiation [77]. The results of these studies proved that PCL/SrBG composite

materials had potential to fabricate the next generation bone scaffolds.

3.5 SCAFFOLD FABRICATION TECHNIQUES

There are several techniques available for the fabrication of 3D composite

scaffolds which feature high porosity and interconnectivity. Currently, the main

fabrication methods used in bone tissue engineering include: additive manufacturing,

electrospinning, thermally-induced phase separation (TIPS), solvent casting/particle

leaching, solid free-form fabrication, microsphere sintering and surface coating of

scaffold [12,50,78]. A summary of advantages and disadvantages of these individual

techniques can be found in Table 3.3 below.

Page 49: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 31

Table 3.3 Comparison of different bone scaffold fabrication techniques [50,66]

Fabrication route Advantages Disadvantages

Thermally induced phase separation (TIPS)

High porosity (~95%)

Highly interconnected pore

structures

Anisotropic and tubular pores

possible

Control of structure and pore size by varying preparation conditions

Long time to sublime solvent

(48 hours)

Shrinkage issues

Small scale production

Use of organic solvents

Solvent casting/particle leaching

Controlled porosity

Controlled interconnectivity (if particles are sintered)

Structures generally

isotropic

Use of organic solvents electrospinning Process versatility and large surface

area to volume ratio

fully interconnected porous structure

Good mechanical properties

Small pore size can be

detrimental for the desired

cell infiltration into the inner

regions [79]

Limit in scaffold thickness

Solid free-form fabrication

Porous structure can be tailored to

host tissue

Protein and cell encapsulation

possible

Good interface with medical imaging

Resolution needs to be

improved to the micro-scale

Some methods use organic solvents

Rapid Prototyping fully interconnected channel network, controlled porosity and channel which mimic the microstructure of living tissue

Relatively expensive and complicated set-up

Microsphere sintering

Graded porosity structures possible

Controlled porosity

Can be fabricated into complex shapes

Interconnectivity is an issue

Use of organic solvents

Scaffold surface coating

Quick and easy Clogging of pores, sometimes organic solvents used, coating adhesion substrate can be too weak

Of these techniques, electrospinning has gained substantial attention with the

bone tissue engineering community in recent years; due to the relatively simple and

low-cost fabrication process, combined with the significant potential to construct bone

Page 50: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

32 Chapter 3: Literature review

scaffolds that mimic native bone matrix [80,81]. To our knowledge, there has been

limited application of electrospun PCL/SrBG composite scaffolds. More details of

electrospinning are introduced in the following sections.

3.5.1 History of electrospinning

Similar to mechanically forced extrusion spinning in textile industries,

electrospinning employs electrostatic forces to produce polymer fibres which range in

diameter from tens of microns down to tens of nanometers [82,83]. First introduced

by Zeleny in 1914, this technique was further developed by Anton Formhals who

repeatedly patented electrospinning for the production of continuous fine fibres for the

textile industry between 1934 and 1944 [82,84]. Electrospinning was not used for

scaffold fabrication in tissue-engineering applications until 1995, when Doshi and

Reneker discovered the potential of using electrospun fibres for tissue engineering

[84,85]. Since then, there has been a strong growth in this area due to the simplicity

and uniqueness of electrospinning. This technique has the ability to produce 3D nano-

fibre scaffolds with large area-volume ratio and interconnected pores with spatial

orientation, which promote cell adherence, proliferation and differentiation [82].

3.5.2 Basic electrospinning equipment

As shown in Figure 3.8, a typical electrospinning setup consists of three major

components: a spinneret, a collector and a high-voltage power source [80,82].

Additionally, a syringe is directly connected to the spinneret as a reservoir for polymer

solution (or melt). To ensure a steady and controllable feed rate, a syringe pump is

employed to serve this purpose.

Page 51: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 33

Figure 3.7 Schematic illustration of typical electrospinning setup (not to scale) [82]

3.5.3 Theory of electrospinning

In the electrospinning process, a high voltage/low current is applied to the

spinneret to create an electrically charged jet of polymer solution or melt. As the

electric field accumulates at the tip of the needle, a pendant droplet of polymer solution

at the spinneret becomes highly electrified which subsequently leads to the elongation

of the hemispherical surface of the droplet to form a shape known as the Taylor cone

[80,82]. The Taylor cone is the result of droplet surface tension and electrostatic force.

Further increasing the electric field will reach a critical value when the repulsive

electrostatic force exceeds the surface tension of the droplet and a fine jet of charged

polymer solution is ejected from the tip of the Taylor cone [80,82,86]. Subjected to

elongation and instabilities, this jet then undergoes a stretching and whipping process,

accompanied by rapid solvent evaporation from the polymer solution, leaving only

charged ultra-fine fibres. Polymer fibres tend to move away from each other due to the

mutual electrostatic repulsion resulting in limited layers of scaffold and a random coil

of nanofibres [80,82,84,86]. In order to obtain desired fibre alignments, various

collector set-ups have been developed over the years, including rotating drum

collectors. A variety of electrospinning configurations were summarised by Sahay et

al. as shown in Figure 3.9 [87].

Page 52: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

34 Chapter 3: Literature review

Figure 3.8 A schematic summary of electrospinning configurations for desired fibre alignments [87]

3.5.4 Process of electrospinning

It is known that the electrospinning process is influenced by polymer solution

properties, process parameters and ambient conditions. From our standpoint, we can

manipulate the variables of this process to fabricate the desired scaffolds. Of these

parameters, the most controllable variables include the flow rate, electric field strength,

distance between spinneret and collector, needle tip design, and collector composition

and geometry [80,86,88]. The influence of individual parameters on the fibre

morphology is summarized in Table 3.4 below.

Table 3.4 Important parameters for electrospinning process [80] Process parameter Effect on fibre morphology

Viscosity/concentration • Low concentration/viscosities yielded defects in the form of beads and junctions; increasing concentration/viscosity reduced the defects

• Fiber diameters increased with increasing concentration/viscosity

Conductivity/solution charge density

• Increasing the conductivity aided in the production of uniform bead-free fibers

Page 53: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 35

• Higher conductivities yielded smaller fibers in general (exceptions were PAA and polyamide-6)

Surface tension • No conclusive link established between surface tension and fiber morphology

Polymer molecular weight • Increasing molecular weight reduced the number of beads and droplets

Dipole moment and dielectric constant

• Successful spinning occurred in solvents with a high dielectric constant

Flow rate • Lower flow rates yielded fibers with smaller diameters

• High flow rates produced fibers that were not dry upon reaching the collector

Distance between tip and collector

• A minimum distance was required to obtain dried fibers

• At distances either too close or too far, beading was observed

Needle tip design • Using a coaxial, 2-caplillary spinneret, hollow fibers were produced

• Multiple needle tips were employed to increase throughput

Collector composition and geometry

• Smoother fibers resulted from metal collectors; more porous fiber structure was obtained using porous collectors

• Aligned fibers were obtained using a conductive frame, rotating drum, or a wheel-like bobbin collector

• Yarns and braided fibers were also obtained

Ambient parameters • Increased temperature caused a decrease in solution viscosity, resulting in smaller fibers

• Increasing humidity resulted in the appearance of circular pores on the fibres

Field strength/voltage • At too high voltage, beading was observed

• Correlation between voltage and fibre diameter was ambiguous

3.5.5 Melt-electrospinning

Generally, there are two ways to prepare liquid polymer: by dissolving the

polymer in a solvent or by melting the polymer. The solution-electrospinning approach

is well established and has been popular for scaffold manufacturing in TE field

compared to melt-electrospinning [86]. However, melt-electrospinning has revealed to

be a potentially more suitable technique for bone scaffold fabrication for the following

reasons. First of all, melt-electrospinning is solvent free while solution-

Page 54: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

36 Chapter 3: Literature review

electrospinning depends on using an organic solution to dissolve the polymer. In most

cases these solvents are toxic, and solution-electrospinning has the potential for trace

harmful solvents remaining in the scaffold which may adversely affect cells and tissues

[89]. Secondly, the melt-electrospinning process is more controllable than solution-

electrospinning attributable to the nature of the process, resulting in better aligned and

thicker fibres. In the solution-electrospinning process, a rapid whipping jet generates

ultrathin fibres within a relatively large collection area often with chaotic deposition

[86,90,91], the subsequent solvent vaporization further decreases the fibre diameter

[80]. According to important results obtained by Mikos et al., larger fibre diameter

facilitates increased pore size and interconnectivity, which in turn promotes cell

infiltration which is an important consideration when we are attempting to make large

tissue engineered scaffolds [80,92]. Thirdly, polymer melts have higher viscosity and

lower conductivity compared to polymer solutions, and there is thus not a need for a

minimum collection distance for melt-electrospinning, which means it is feasible to

collect polymer jets in the stability region, enabling fabrication of scaffolds of ordered

layout. While for solution-electrospinning, it is really necessary to maintain the

distance between the spinneret and collector to allow solvent removal by evaporation,

leading to random meshes of polymer jets collected in the instability region [90]. These

unique properties of melt-electrospinning have enabled the emergence of a new

biofabrication technique: melt-electrospinning writing (MEW) [93]. MEW uses

additive manufacturing principles that allow the layer-by-layer fibre deposition like

FDM to fabricate scaffolds with specific designs, shapes, porosities and sizes [93,94].

MEW is realised by moving the collector in x, y and z coordinates according to pre-

programmed patterns allowing the polymer jet to deposit into coherent 3D structures,

which makes it possible to produce scaffolds tailored to patients’ specific defect size.

3.5.6 Hybrid electrospinning system

Although rarely investigated, several studies have reported scaffold fabrication

processed that combine usage of solvents and elevated temperature, which is termed

as hybrid systems of melt- and solution-electrospinning [83]. Different to other

‘hybrid’ systems which are simply to combine the electrospun fibres produced from

individual solution- and melt-electrospinning [95], these hybrid systems defined by

Hutmacher and Dalton actually merges the two electrospinning processes to dissolve

the polymers in solvents at elevated temperature. These ‘melt’ assisted solution-

Page 55: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 37

electrospinning systems have shown better solvent removal during the process via

accelerated evaporation and improved polymer solubility in the solvents, and also

possibility to take advantage of physical transformations due to thermal changes [96].

Some polymers, such as polyethylene (PE), are not considered amenable to

electrospinning as it readily crystallises from solution due to its low solubility. In the

study by Givens et al., the authors successfully produced polyethylene microfibers via

conventional solution-electrospinning (2-5 wt%) by maintaining the electrospinning

temperature at 105-110 °C with an IR emitter system [97]. Yoshioka et al. then adopted

the same hybrid solution-electrospinning system with improved temperature control

to produce PE electrospun fibres in nano scale [98]. The hybrid electrospinning system

shows the capacity to overcome some of the limitations of materials selection

experienced with individual solution-/melt-electrospinning. These systems could

potentially further advance the TE field by expanding the spectrum of materials for

scaffolds production, including polymer/bioceramic composite biomaterials

conventionally not successful.

3.6 TE SCAFFOLDS ASSESSMENT

As a result of our aging population and decreased physical activity levels, the

annual market for bone grafts is in excess of $2.5 billion and is expected to further

grow at 7-8% every year [99]. This demand presents great needs and potential growth

for TE strategies. However, before translating the TE scaffolds into clinical

applications, a series of bio-capability tests must be undertaken in vitro and in vivo

[100]. In vitro testing uses isolated cells for the characterisation of biological activity

effects of bone TE scaffolds and for screening acute toxicity and cytocompatibility. In

vitro assays are considered the first stage testing and are popular as they also give

information regarding how well cells attach, proliferate and differentiate on the

scaffolds [101]. The information obtained from in vitro studies serves as a valuable

guide for the following in vivo tests and also minimise animal usage. However, in vitro

cell culture does not reflect the complex in vivo environments of implanted scaffolds,

and specifically for a bone implant. It lacks control of physiological loading which has

proven to be essential for bone development [101]. In comparison, in vivo testing uses

animal models to evaluate scaffolds in a reproducible approximation of the mechanical

and physiological clinical situation [100,101]. For this purpose, a number of animal

models have been established, ranging from small animals such as a mouse to large

Page 56: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

38 Chapter 3: Literature review

animals such as sheep [100]. The small animal models have shown fast bone turnover

rate, cheaper maintenance and are easier to standardise, but they do not closely mimic

human bone architecture [100]. On the other hand, the large animal models have

shown a closer resemblance to human bone architecture, physiology and

biomechanical properties [100]. Their disadvantages include expensive maintenance

cost, lower bone turnover and difficulties in standardisation. Given their limitations,

these animal models are actively used in the TE field where appropriate [102–105].

For the accurate interpretation of these in vivo test results, comprehensive post

explantation analysis techniques are employed, including micro-computed

tomography, histology, scanning electron microscopy and mechanical testing

[105,106]. Among these ex vivo tests, histology is a key assessment to characterise

bone formation and mineralisation mechanisms including tissue, cellular and

molecular pattern and distribution within the bone matrix. In addition, histology also

provides information on the nature of soft tissues and scaffold/implant interface with

mineralised bone [105]. Immunohistochemistry (IHC) is a vital histological technique

that can further characterise osteogenic/angiogenic markers presented on the

regenerated bone tissue [104,107]. IHC demonstrates specific markers on the surface

of tissue through antibody-antigen interaction either directly or indirectly [108].

Commonly the antibody is conjugated a fluorescent chemical or an enzyme (normally

a peroxidase) that can convert a substrate to a visible dye, the antibody distribution is

assumed to reflect antigen location [108].

3.6.1 Histology

Histology, which is essential for the understanding of disciplines including

biology, medicine and veterinary medicine, is the scientific study of the fine detail of

cells and tissues to study their structure and composition at the microscopic level [6].

It provides microscopic analysis of two-dimensional thin tissue slice (called a ‘section’)

carefully prepared using special subsequent procedures of tissue fixation, dehydration,

infiltration and embedding, followed by sectioning and staining [109]. Because

biological tissues/TE scaffolds must be supported in a hard matrix to allow sufficiently

thin sections to be cut, the tissue dehydration and infiltration process are necessary to

remove and then replace water within tissues with a medium that solidifies to allow

thin sectioning [109]. Histology is a highly versatile technique, numerous stains are

available for specific tissue types, and it is used in routine medical analysis and is now

Page 57: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 39

widely employed in tissue engineering research for the ex vivo analysis [105]. Besides

conventional stains, immunohistochemistry (IHC) and enzyme activity assays, such as

tartrate-resistant acid phosphatase (TRAP) and alkaline phosphatase (ALP) staining

can detect specific antigens (e.g. proteins) and enzymes providing a high degree of

details of tissue composition unmatched by other techniques [6,110].

Paraffin processing is the most routinely used tissue preparation technique for

histology [109]. In this process, the water content of the specimen is removed through

a series of dehydration steps, and then the specimen is impregnated with paraffin to

create a more homogeneous cutting profile. The paraffin embedded specimens are

sectioned via a microtome, producing sections between 2 and 10 microns in thickness

[109]. Due to the ease of use, high throughput and efficiency, long storage life, and

high quality final results, the paraffin route has become the most commonly used

preparation technique. A broad variety of tissue types and species have near identical

paraffin processing parameters, making it an extremely robust and versatile technique.

However, the sample sizes are limited by the embedding mould to 3.5×2.2×1 cm in

length, width and depth respectively.

Calcified tissues, such as bone and tooth, are an exception and are not readily

suited to paraffin processing [111]. The dense mineralised component of calcified

tissues makes them too hard for the standard sectioning process, mainly because of the

mismatch of hardness and density between bone tissue and paraffin embedding media,

leading to the dissatisfactory quality of the produced section. To tackle this problem a

variety of embedding and sectioning techniques designed specifically for calcified

tissues have been developed and used for over three decades [109,110,112,113]. These

techniques can be summarised into two distinct approaches: 1) decalcification of the

mineral component of the tissue, followed by paraffin processing; 2) and resin

embedding for fully calcified samples [111,114].

Paraffin approach

In order to section bone tissue with a standard paraffin microtome, the calcium

salts within the tissue must be removed by a process called ‘decalcification’ before

paraffin embedding [113]. Decalcification is a necessary step to produce high quality

paraffin sections of bone specimens [6,109]. Routine decalcification methods use

chemical reagents ranging from slow ‘gentle’ metal ion chelating agents such as

ethylenediaminetetraacetic acid (EDTA) [109,113,115] to ‘harsh’ rapid acting acid

Page 58: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

40 Chapter 3: Literature review

solutions such as hydrochloric acid [116,117]. The time taken to decalcify bone

specimens varies significantly dependent upon the size of the sample being decalcified,

the type of decalcification solution and temperature [113,118]. Acid solutions and

higher temperatures yield more rapid decalcification, but this can come at the cost of

reduced preservation of tissue/cell structure and poor enzyme and epitope preservation

or even complete digestion of bone, leading to inferior quality final results [113].

Decalcification of tissue in EDTA at 4oC has been shown to be effective in preserving

structure and epitope, however, it is a slow process which can take weeks to complete

dependent upon sample size and initial mineral content [119]. The decalcification

process enables the production of paraffin sections of bone and other calcified tissues,

however, it has two major disadvantages: potentially poor preservation of bone matrix

structure [110] and the inability to assess mineralisation of the bone tissue due to the

removal of the calcified phase [113]. As such, alternative approaches have been

developed which negate the need to decalcify the bone specimens at all: resin

embedding.

Resin approach

This technique is the ‘gold standard’ for undecalcified bone and metallic implant

analysis. The procedures of resin embedding have much in common with routine

paraffin approach, including i) fixation, ii) dehydration, iii) clearing, iv) infiltration, v)

resin embedding and vi) sectioning [113]. However, compared to paraffin section

preparation, resin approaches do not require decalcification as resin as an embedding

media can better match the hardness of bone specimens than paraffin, which allows

for a greater preservation of tissue morphology as well as an assessment of the mineral

content of the bone tissue. Many resins have been used, ranging from araldites [120]

to acrylics [121] with methyl methacrylate (MMA) based resins being most commonly

used [110]. MMA has a long history of use in hard tissue research spanning almost 50

years [8,110,122]. One great drawback of MMA embedding media is the high

polymerisation temperatures (~80 °C) as a result of the exothermic reaction [121]. The

heat generated during the embedding process initiates the destruction of epitopes and

enzymes of bone tissues, therefore, MMA embedding is considered to be unsuitable

for specimens which require IHC or enzyme analysis [121]. Over the last few decades,

low temperature polymerizing resin have been developed to overcome this limitation

[8,110]. Technovit 9100 New® resin, a low temperature curing MMA-based resin, has

Page 59: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 41

become commercially available since 2000 [110]. Technovit 9100 New® has the

capacity to polymerise in an environment as low as -20 °C allowing the preservation

of epitopes, which makes it suitable for IHC assessments [8,110,121,123]

Resin embedded specimens can be sectioned by a microtome [104,106,122,124–

128] using a heavy duty blade or a purpose built resin microtome, producing sections

approximately 8 to 10 microns thick. The other routine section technique is ground

sectioning where specimen resin blocks are cut thicker (200 microns) with a saw blade

and then ground down 80 to 30 microns prior to staining [111,129–135]. The ground

sectioning technique for resin sections is more robust than resin microtome sectioning,

as it also allows for sectioning of a wider variety of calcified tissue and implants

including metallic implants in cortical bone in situ [136]. This technique is able to

provide a detailed analysis of tissue-implant interactions unattainable by other classical

histological approaches as they require the removal of the implant prior to embedding

and sectioning [8,135]. One major drawback of the ground sectioning technique is the

large consumption of specimen, taking approximately 1 mm of specimen per produced

section compared to several microns in microtome sectioning. It’s also highly technical

and time-consuming.

Histological stains

When focusing on histology of bone, most of the routine histological stains have

been developed for paraffin embedded samples. The most commonly utilised ones are

haematoxylin and eosin (H&E) [114] and IHC [109]. While H&E is an effective and

simple general stain, it has limited capacity to differentiate mineralised hard tissues

from surrounding tissues by colour, which is not ideal for tissue engineers to determine

bone regeneration. In order to improve this situation, stains such as von Kossa silver

nitrate methods and Goldner’s trichrome staining were developed and employed [8].

Goldner’s trichrome is an effective staining technique employed in numerous in vivo

bone regeneration studies, this stain provides comprehensive detail of calcified tissues

(typically in green colour), with strong contrasting colours [104,106,121,124–

135,137–139] as well as decalcified ones (red) and connective tissues (orange) [140].

In this thesis, I sought to compare and contrast various histological methods for the

purpose of bone TE applications.

Page 60: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

42 Chapter 3: Literature review

3.7 CONCLUSION

Over the last two decades, enormous effort has been made in developing the

ideal bone substitute materials and a broad variety of materials have been investigated

by researchers around the world. To date, composite materials comprising

biodegradable polymers and bioactive glasses are promising candidates, as they meet

most of the requirements for bone regenerative scaffolds. PCL/SrBG composites will

be the primary material for this research project due to their excellent individual

properties which complement each other when combined. These composite scaffolds

are potentially bioresorbable and bioactive and capable of supporting osteoblasts

adhesion, spreading and viability.

To make this composite material into a viable bone scaffold, optimal

manufacturing techniques are vital. Electrospinning has been established as an easy-

to-operate and low-cost scaffold fabrication technique, and its popularity with the bone

tissue engineering community rocketed in the past decade, demonstrated by a large

number of publications. By controlling the key parameters of the electrospinning

process, we can fabricate bone scaffolds with desired size, shape and mechanical

strength to fit in the defect site. As described above, melt-electrospinning provides us

with larger and thicker solvent-free scaffolds with better mechanical properties

compared to scaffolds produced using solution-electrospinning. In the context of this

project, as the scaffolds may eventually be applied in low load-bearing conditions, it

is necessary for them to be structurally sound with adequate mechanical strength.

Therefore, melt-electrospinning is the preferable scaffold fabrication technique for this

purpose.

By combining the advantages of the composite PCL/SrBG with the melt-

electrospinning process, we are able to fabricate promising bone scaffolds that are

osteoconductive and osteoinductive, and with appropriate mechanical properties.

Bone TE research demands rigorous characterisation methods to examine the

progress of bone regeneration in the defect sites via in vivo studies, which are pivotal

to provide a complete understanding of the bone healing capacity of the materials and

scaffolds. However, in vivo studies are considerably resource consuming. Considering

that numbers of animals that can be used in these studies are also limited, the samples

obtained from these in vivo studies are extremely precious, and the data achieved from

such samples need to be rigorous and relevant. It still remains a challenge to obtain

Page 61: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 3: Literature review 43

maximal information of bone specimens [8], thus, it is imperative to continuously

optimise and improve the current histological techniques to meet the demand of

processing more complex and large bone/implant explants, and to ensure that bone TE

strategies have every opportunity to reach the clinic to treat patient suffering tissue

loss.

Page 62: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 63: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 45

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

This study has been completed and published.

Jiongyu Ren, et al. Melt-electrospun polycaprolactone strontium-substituted

bioactive glass scaffolds for bone regeneration. J. Biomed. Mater. Res. - Part A 102,

3140–3153 (2014)

4.1 INTRODUCTION

Electrospinning is a versatile technique for the production of fibres ranging from

the micro to nano scale in diameter [82]. Many different polymers may be electrospun

via two different techniques: dissolving the polymer in an organic solvent (solution-

electrospinning) [141], and melting the polymer (melt-electrospinning) [142]. Melt-

electrospinning overcomes technical restrictions governed by solvent accumulation

and possible toxicity to cells [90]. In spite of the potential benefits of melt-

electrospinning, it has not yet gained widespread attention in the field of tissue

engineering (TE) when compared to solution-electrospinning.

Bone TE aims to combine aspects of conventional biology, engineering,

medicine, and chemistry to produce functional scaffolds for clinical treatments of bone

loss [143]. Current treatments in clinical practice rely on bone grafting, the gold

standard of which is autografting which involves taking bone tissue from a donor site

on the same patient. This technique has prominent issues related to limited supply,

donor site morbidity and surgical complications [28] hence it is important to develop

alternative solutions and move towards off-the-shelf solutions for bone loss.

Developing TE scaffolds to be used for the treatment of bone defects presents

challenges, including those associated with the need for a porous structure to allow for

good cellular infiltration and vascular ingrowth, while still maintaining the required

mechanical strength to support the tissue [144]. These scaffolds are commonly

constructed from either synthetic or natural biodegradable polymeric materials.

Page 64: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

46 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Polycaprolactone (PCL) belongs to the family of aliphatic polyesters and is

becoming an increasingly popular FDA-approved biomaterial [45]. PCL can be easily

fabricated into a scaffold of almost any size and shape by a variety of fabrication

techniques, owing to its low melting point (60 °C), and superior rheological and

viscoelastic properties compared to other synthetic polymers. These properties make

melt-electrospinning of PCL easily achievable; in contrast, more commonly used

polyesters, such as poly(L) lactic acid (PLLA), require temperatures in excess of 200

°C for successful melt-electrospinning [145]. Although PCL has many desirable

features, making it a promising candidate for a bone scaffold material, it is limited by

its hydrophobicity which is not conducive to cell attachment [33,46] and lack of

osteoconductivity [146]. In order to improve the suitability of PCL scaffolds for bone

TE applications, composite systems comprising the biodegradable polymer matrix

combined with inorganic components are emerging as promising candidates. Early

research into developing composite scaffolds has focused upon improving the

osteoinductive potential of the polymeric component by the incorporation of a

biocompatible inorganic phase such as hydroxyapatite (HA), ceramics and bioactive

glasses [50,72,147].

Bioactive glasses (BGs) are a group of inorganic bioactive materials that have

been shown to form a strong bond with hard and soft tissues; a result of carbonate-

substituted hydroxyapatite layer formation at the interface [51,148]. The first

commercially available BG was introduced in the 1970s, named 45S5, (commercial

name, Bioglass®), with a chemical composition of SiO2-P2O5-Na2O-CaO. Since then,

a large variety of BGs based upon derivations of the 45S5 composition have been

developed and applied in bone TE [48,149,150].

Gentleman et al. have developed strontium-substituted BG (SrBG) by

substituting 0-100% of the calcium (Ca) component of the 45S5 formulation with

strontium (Sr) [58]. It has already been established that Sr ions may significantly

enhance bone regeneration [49,57,59], and Sr-dosed drugs such as Sr-ranelate,

marketed under the trade name Protelos™, are available on the market for treating

osteoporosis [55,56]. In vitro results have shown that SrBG not only promotes

osteoblast proliferation and alkaline phosphatase (ALP) activity when directly applied

to the human osteosarcoma cell line, Saos-2, but also inhibits osteoclast differentiation

Page 65: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 47

and decreases tartrate resistant acid phosphatase (TRAP) activity in the mouse

monocyte cell line, RAW264.7 [56,58].

To our knowledge, the production of composite scaffolds comprising PCL and

SrBG utilising the technique of melt-electrospinning has not been achieved to-date.

Here I report for the first time the ability to produce composite scaffolds suitable for

bone TE applications using melt-electrospinning approaches. I show PCL/SrBG

scaffolds with interconnective pores, structural robustness, and the potential to

promote osteogenesis. In this initial study, I firstly optimised techniques for melt-

electrospinning PCL fibres with SrBG particles homogeneously dispersed throughout

the fibres. I furthermore assessed the scaffolds’ ability to support cell attachment and

proliferation in addition to the osteogenic potential of the composite scaffold in vitro.

4.2 MATERIALS AND METHODS

4.2.1 Scaffold fabrication and characterisation

PCL/SrBG composite preparation and scaffold fabrication

The 75% strontium-substituted bioactive glass (SrBG) was produced by Stevens

et al as described in [57,151]. Briefly, the composition of BG containing 46.13 SiO2 –

2.60 P2O5 – 24.35 Na2O – 26.91 (SrO:CaO) (mole %) where 75% of the calcium was

substituted with strontium was melted in a platinum crucible at 1400 °C for 90 min

and rapidly quenched into deionised water to form frits. SrBG frits were then ground

and sieved to yield particles < 38 μm in diameter. In order to obtain a homogeneous

distribution of SrBG within the Polycaprolactone (PCL, Capa 6400, Perstorp UK

Limited), PCL was first melted on a hotplate (~70 °C). SrBG particles (10% wt.) were

then added and manually mixed into the PCL melt for 20 mins before being loaded

into a 2 ml syringe. Air bubbles were removed from the composite melt after loading

into the syringe via placement of the syringe in a vacuum oven (-80 kPa for 30 mins

at 90 °C).

Both PCL and PCL/SrBG composite scaffolds were produced using the melt-

electrospinning technique as previously described [90], using the optimised

parameters (Table 4.1).

Page 66: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

48 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Table 4.1 Parameters for melt-electrospun scaffold fabrication Scaffold type Voltage Temperature Collection

distance Needle gauge

Flow rate

PCL 7 KV 80 °C 4 cm G21 20 μl/h

PCL/SrBG 7 KV 85 °C 6 cm G19 20 μl/h

Scaffold surface modification via sodium hydroxide (NaOH) etching was

performed to reduce the hydrophobicity of PCL, increase the surface roughness of the

fibres and to increase surface exposure of SrBG particles to facilitate enhanced cell

attachment [88]. As reported in the literature [47], scaffolds were etched with 5M

NaOH for 1 hour @ 37 °C and subsequently washed with dH2O until the supernatant

pH reached 7.

Scaffold characterization

Light microscopy PCL and PCL/SrBG scaffolds were weighed with an

AUW2200 analytical balance (Shimadzu, Canada); Light microscopy (Zeiss Axio

Imager A2, Germany) and a Quanta 200 scanning electron microscope (SEM, FEI

Australia, Australia) were used to assess morphology and diameter of the fibres were

assessed using ZEN blue software (Zeiss, Germany). Average fibre diameters were

determined by random selection of 6 scaffolds per group. A total of 42 fibres per group

were measured via the ZEN imaging software.

Micro-computed tomography (µCT) PCL and PCL/SrBG (n = 2) scaffolds

were scanned in the air in a micro-computed tomography scanner (µCT, 40Scanco

Medical, Brüttisellen, Switzerland), at an energy of 45 kVp and intensity of 177 µA

with 300 ms integration time. The scans were reconstructed to three-dimensional

datasets with an isotropic voxel size of 6 µm. The scans were segmented to visualise

bioglass particle distribution throughout the scaffolds. Next, the scans were analysed

with the scanner’s software using the distance transformation method to determine

interfibre spacing.

4.2.2 Ion dissolution and precipitation

PCL and PCL/SrBG scaffolds were submerged in alpha-Minimum Essential

Medium (α-MEM, Invitrogen, Australia) at a ratio of 5 g/L supplemented with 1%

(v/v) of penicillin-streptomycin (Invitrogen, Australia), incubated at 37 °C/5% CO2.

Page 67: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 49

Samples of dissolution ion media were collected after 6 hours, 1, 2, 3, 7, 14, 21 and 28

days for elemental analysis.

Elemental concentrations of calcium (Ca), phosphorus (P), silicon (Si) and

strontium (Sr) were measured on a Vista MPX Inductively Coupled Plasma - Optical

Emission Spectrometer (ICP-OES, Varian, USA).

In order to identify elemental deposition on the surface of the scaffolds, energy-

dispersive X-ray spectroscopy (EDX) (Quanta 200, FEI, Australia) was used to scan

the surfaces of PCL and PCL/SrBG fibres.

4.2.3 In vitro studies

To examine the osteogenic potential of the PCL/SrBG composite scaffolds, cell

culture experiments which considered cell metabolic activity, proliferation, viability

and gene expression upon the scaffolds were undertaken using mouse osteoblast

precursor cell line, MC3T3-E1 (passage 9).

Cell culture

Prior to experimentation, MC3T3 cells (sub-clone 14) [152] were cultured in

growth media: α-MEM cell culture media supplemented with 10% (v/v) foetal bovine

serum (FBS, Invitrogen, Australia) and 1% (v/v) penicillin-streptomycin at 37 °C/5%

CO2. Scaffolds were cut with a 6 mm biopsy punch and sterilised with 70% ethanol,

followed by UV irradiation for 20 min each side. MC3T3 cells were seeded at a density

of 4500 cells per scaffold (159 cells/mm2), after 24 hours culture, half the samples

were treated with osteogenic media (growth media supplemented with 10mM β-

glycerophosphate, 50 µg/ml ascorbic acid and 0.1mM dexamethasone (all from

Sigma)), the other half were cultured in growth media as a control. A total of four

different groups were studied as shown in Table 4.2 for the following experiments.

Table 4.2 In vitro experimental groups of study 1. PCL/SrBG scaffolds (10 wt%) were studied in both growth media and osteogenic media and PCL scaffolds were studied as control.

Group Scaffold type Culture media type n

1. PCL_C PCL Growth (control) 164

2. PCL_O PCL Osteogenic 164

3. PCL/SrBG_C PCL/SrBG Growth (control) 164

4. PCL/SrBG_O PCL/SrBG Osteogenic 164

Page 68: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

50 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

LIVE/DEAD staining

A qualitative LIVE/DEAD assay was used to indicate both the cell viability and

cell distribution within the seeded scaffolds [153]. After days 1 and 7, samples were

moved into a fresh plate, cell culture media was removed and the scaffolds were

washed twice with phosphate buffered saline (PBS, Invitrogen). The scaffolds were

then incubated with 0.67 µg/ml fluorescein diacetate (FDA, Invitrogen) and 5 µg/ml

propidium iodide (PI, Invitrogen) for 5 min in the dark. The staining was visualized

using a Zeiss Axio M2 Imager (Zeiss, Germany) fluorescent microscope (at λ = 488

nm and λ = 568 nm excitation).

Assessment of cell attachment, morphology, and BG identification

Scanning Electron Microscopy (SEM) was used to qualitatively investigate

cellular attachment and morphology on the respective scaffolds. As described

previously [154], after days 3, 7, 14, 21 and 28, scaffolds were fixed in 3%

glutaraldehyde immediately following cell culture. Scaffolds collected from all time

points were washed in sodium cacodylate buffer (Sigma, Australia) and 1% osmium

tetroxides in cacodylate. Samples were then washed twice in ultrapure UHQ water

(Millipore Australia, Australia) and dehydrated in a graded ethanol followed by

Hexamethyldisilazane drying (all reagents were supplied by ProSciTech, Australia).

After sputter coating with gold, samples were visualized using a Quanta 200 SEM

(FEI, Australia).

Confocal Laser Scanning Microscopy (CLSM) [155] was used to visualise the

morphology of actin fibres and nuclei of MC3T3 cells on the scaffolds. Here we

introduce a new way to visualise the cell attachment onto SrBG particles by combining

actin staining and nuclei staining with Alizarin red S staining (to stain SrBG particles).

Briefly, after 3, 7, 14, 21, 28 days, cell culture media were removed and scaffolds were

transferred into fresh 48-well plates. After 2 careful washes with PBS supplemented

with 0.5 mM Mg2+, 0.9 mM Ca2+, scaffolds were fixed with 4% paraformaldehyde

(PFA, Sigma) solution for 30 min at room temperature. Samples were then washed

with PBS and permeabilized with 0.2% (v/v) Triton X-100 (Invitrogen)/PBS solution

for 5 min, followed by 2 washes with PBS. Samples were then incubated with 0.5%

Page 69: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 51

(w/v) Bovine serum albumin (BSA, Sigma)/PBS for 10 min, followed by 0.5% (v/v)

BSA/PBS solution containing 0.8 U/ml Alexa Fluor® 488 Phalloidin (Invitrogen) and

5 µg/ml 4’,6-diamino-2-phenylindole (DAPI, Invitrogen). After 1 wash with MiliQ

water, the scaffolds were stained with Alizarin red S (pH 4.2) for 5 min and washed

with MiliQ water twice to remove excess stain. The scaffolds were then stored in PBS

until imaging. The PCL/SrBG scaffolds were visualized with Leica SP5 Confocal

microscope (Leica, Germany), images of identically treated PCL only scaffolds were

taken as controls.

Cell metabolic assay

MTT (3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; Sigma,

Australia) is a cell metabolic activity assay which works on the principle of measuring

the absorbance of formazan, which is reduced from MTT by mitochondria in active

cells [58]. This was used to assess the metabolic activity of the cells when cultured on

the different scaffolds with and without the presence of osteogenic inducing media. At

days 1, 7, 14, 21 and 28, scaffolds (n = 6) were transferred into a fresh 48-well plate

and 500 µl of fresh media supplemented with 20 µl of MTT solution (5 mg/ml) was

added. Scaffolds were incubated (37 °C/5% CO2) for 4 hours, after which the media

was removed and 100 µl (D1, 7), 200 µl (D14), 400 µl (D21) or 500 µl (D28) dimethyl

sulfoxide (DMSO, Merck, Australia) was added to each well. The plates were then

covered with tinfoil and placed on an orbital shaker for 10 min before 100 µl of DMSO

eluant was taken from each well and transferred into fresh 96-well plates and

absorption at λ = 540 nm was measured. The obtained reading was multiplied with the

dilution factors of DMSO at all 5 time points respectively.

Alizarin red S staining (mineralisation)

Mineralisation of the cells on the scaffold was used as an indicator of

differentiation of the MC3T3 cells. I used a qualitative detection based on the selective

bonding between Alizarin red S and calcium salts [156]. After 7, 14, 21 and 28 days,

cell culture media was removed from the well and scaffolds (n = 6) were first washed

in PBS and then fixed with ice-cold methanol. Alizarin red S (Sigma, Australia) dye

solution (1 g of powder into 50 ml of distilled water) of 150 µl was added to each

scaffold. The pH of Alizarin red S solution was maintained at 4.15 for all time points.

Page 70: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

52 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Scaffolds were incubated for 5 min and then were washed repeatedly with MiliQ water

until the solution ran clear. Plates were scanned via a Canoscan 8600F flatbed scanner

(Cannon, US).

Cell differentiation

In order to compare the osteogenic potential of the cells cultured on PCL/SrBG

scaffolds to those on PCL scaffolds, I measured Alkaline Phosphatase (ALP) activities

of the cells adhere on each scaffold. ALP is a known early marker of osteoblast

differentiation and plays a key role in mineralisation [58]. In order to represent the

ALP activity as activity per cell, I normalized the ALP data with a total DNA

quantification technique (the PicoGreen assay) described hereafter.

For both assays, at the 7, 14, 21 and 28 day time points, cell culture media was

removed and scaffolds were washed twice with PBS and transferred into fresh 48-well

plates. 300 μl of 0.2 Triton-X/ ×1 TE buffer was added to each well and samples were

stored at -80 °C until further processing. Scaffolds were scraped with pipettes until

cells were adequately lysed, followed by sonication. Cell lysates were transferred into

1.7 ml Eppendorf tubes. From this point, lysates were assayed for DNA content and

ALP activity.

ALP assay

100 μl of cell lysate was transferred into a 96-well transparent microplate in

triplicates. 200 μl of para-Nitrophenyl phosphate (pNPP, Sigma, Australia) was added

into each well containing lysate or Triton-X/ ×1 TE buffer as blanks. The plates were

incubated in the dark for precisely 30 min at room temperature on a rocker plate. The

absorbance was read on a BIO-RAD multi-well plate reader at wavelength = 405 nm

(n = 6).

PicoGreen assay

PicoGreen is a cyanine dye that selectively binds dsDNA and sensitively detects

the DNA content which reflects cell numbers, as such, this assay may be used to

indicate cell proliferation [157]. Here I used PicoGreen to quantify the total amount of

DNA and then converted it to a cell number using a conversion factor of 8 pg of DNA

per MC3T3 cell [158]. The PicoGreen assay (Invitrogen, Australia) was performed

according to the manufacturer’s instructions. Briefly, samples were diluted into 40:60,

Page 71: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 53

20:80 and 10:90 (lysate: ×1 TE buffer) in fresh 96-well plates. After the solution was

mixed, 100 μl of ×1 PicoGreen dye solution were added to each sample, making a total

volume of 200 μl in each well. The 96-well plates were covered with aluminium foil

and incubated at room temperature for precisely 5 min on a rocker plate. The plates

were then read by OPTIMA plate reader (BMG LABTECH, Australia) at excitation λ

= 480 nm and emission λ = 520 nm (n = 6).

The ALP absorbance obtained from the assay was normalised by dividing by the

cell number to give the value of ALP activity per cell. The cell number was obtained

from the DNA quantity via the PicoGreen assay.

Real-time quantitative polymerase chain reaction (RT-qPCR)

In order to obtain more detailed information about the osteogenic effect the

SrBG component of the scaffolds has upon the MC3T3 cells, RT-qPCR was used to

quantify the up-regulation of specific genes associated with early stage osteogenesis

[159].

After days 7, 14, 21 and 28, cell culture media was removed and scaffolds (n =

9) were transferred into fresh 48-well plates with 3 scaffolds per well. 300 μl of lysis

buffer containing 1% (v/v) mercaptoethanol (Sigma, Australia) and 7 μl of carrier

RNA (5 ng/μl) was added to each well. RNA was isolated with PureLink RNA Micro

Kit (Invitrogen, Australia) according to the manufacturer’s instructions.

The total quantity and purity of the extracted RNA were tested with a Nanodrop

Microvolume UV-Vis spectrophotometer (ThermoFisher Scientific, Australia). The

concentration of RNA was calculated to obtain a total of 1000 ng cDNA per sample.

The reverse transcription process was carried out with DyNAmo* cDNA Synthesis

Kit (Thermo Scientific, Australia) following manufacturer’s instructions.

RT-qPCR was performed on an ABI Prism 7000 Thermal Cycler (Applied

Biosystems, Australia) using SYBR Green as detection reagent following the same

procedure that has been reported previously [159]. The relative mRNA expressions of

ALP and Osteocalcin (OCN) were assayed and normalized against the house keeping

gene β-ACTIN. Each sample was analysed in triplicate. The mean cycle threshold (Ct)

of each target gene was normalized against Ct of β-ACTIN; the relative expression

calculated using the following formula: 2-(normalized average Cts) ×104.

Page 72: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

54 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Collagen staining in extracellular matrix (ECM)

Van Gieson’ stain (Dorn & Hart Microedge Inc, USA) was used to stain collagen

deposited on the scaffolds during cell culture. After 7, 14, 21 and 28 days, samples (n

= 3) were washed twice in PBS, transferred into a fresh 48-well plate and fixed in 10%

neutral buffered formalin (NBF, Sigma, Australia). Samples were then washed once

with PBS, followed by two washes with MiliQ water and then stained with 150 μl of

Van Gieson per scaffold for 20 min. Samples were rinsed with MiliQ water until the

solution ran clear. The plate was scanned and light microscope images were taken with

a Zeiss Axio Imager M2 microscope (Zeiss, Germany).

4.2.4 Statistical analyses

Statistical significance between groups in MTT, ALP and RT-qPCR assays was

assessed by Two-way ANOVA with a post-hoc Tukey test using IBM SPSS Statistics

Software (Version 19). A p value of less than 0.05 was considered statistically

significant.

4.3 RESULTS

4.3.1 Characterization of PCL and PCL/SrBG scaffolds

PCL and PCL/SrBG were successfully electrospun into fibrous mats. Light

microscopy indicated that the two scaffold types had similar morphologies (Figure

4.1); however, the PCL/SrBG fibres (46.1 +/- 16.6 µm) were slightly larger in diameter

than the PCL fibres (30.6 +/- 1.8 µm) (n = 42, p < 0.05). The µCT analysis determined

the average fibre spacing within the scaffold was 135.6 +/- 64.3 µm for the PCL fibres

and 222.1 +/- 113.6 µm for the PCL/SrBG ones. µCT analysis demonstrated that the

SrBG particles were homogeneously distributed throughout the PCL fibres (Figure

4.1f). No significant difference was detected between PCL scaffolds and PCL/SrBG

scaffolds in terms of weight (n = 70, p > 0.05).

Page 73: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 55

Figure 4.1 Electrospun scaffolds light microscopy and microCT characterization. Light microscopy images of melt-electrospun PCL lower magnification (a) and higher magnification (c) and melt-electrospun PCL/SrBG lower magnification (b) and higher magnification (d), scale bars are as shown. μCT image of PCL scaffold shown as a control (e). PCL/SrBG scaffold showing the distribution of bioactive glass particles (highlighted in red) throughout the scaffold (f), a higher magnification image is shown in (h). Image (g) shows the SrBG particles only.

4.3.2 Ion dissolution and precipitation analysis

EDX analysis indicated the presence of elements Ca and P on the surface of

PCL/SrBG scaffold, and this calcium phosphate (CaP) layer was also visible under

SEM (Figure 4.2a, shows an example image that was taken after 14 days incubation in

Page 74: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

56 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

α-MEM at 37oC). No Ca or P was found on the surface of the PCL only scaffolds at

the same time point (Figure 4.2b). The elemental concentrations of calcium,

phosphorus, silicon and strontium released from both PCL and PCL/SrBG scaffolds

into cell culture media are shown in Figure 4.2(c, d, e and f). In the PCL/SrBG group,

the calcium and phosphate concentrations (in the media) declined rapidly after 3 days

in cell culture, and then further decreased over the next 3 weeks. No significant change

in calcium and phosphate concentrations was observed in cell culture media exposed

to PCL only scaffolds. In the PCL/SrBG group, the concentrations of both silicon and

strontium increased rapidly after 3 days in cell culture and were stable after 14 days in

cell culture. No changes in silicon and strontium concentrations were observed in PCL

only groups.

Page 75: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 57

Figure 4.2 EDX image of melt-electrospun scaffolds after 14 days incubation in α-MEM. (a) PCL/SrBG scaffolds showing a deposition of CaP (Calcium phosphate layer) over the surface of the fibres and (b) PCL scaffolds showing no CaP deposition on the surface of the fibres, scale bar as shown. Elemental analysis of the concentrations of Ca (c), P (d), Si (e) and Sr (f) in α-MEM over 28 days as determined by ICP-OES testing.

4.3.3 In vitro studies

Attachment and proliferation of MC3T3 cells on scaffolds

The LIVE/DEAD staining showed viable MC3T3 cells (as shown in green)

evenly distributed across both PCL and PCL/SrBG scaffolds after 1 day cell culture

with negligible red stained cells detected. After 7 days, the cell number had

significantly increased (Figure 4.3). High magnification images obtained by SEM

showed cells bridging to adjacent fibres on scaffolds in all four groups (Figure 4.4).

Page 76: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

58 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

CLSM images showed numerous filopodia of MC3T3 cells attaching onto the SrBG

particles exposed on PCL/SrBG fibre surfaces (Figure 4.5a) whereas cells attached and

spread evenly on PCL fibres (Figure 4.5b). The MTT assay indicated that the cell

metabolic activity increased with length of cell culture time and significantly increased

after 7 days and 14 days in culture in all four experimental groups (Figure 4.6).

Figure 4.3 LIVE/DEAD staining of MC3T3 cells cultured on melt-electrospun scaffolds. (a) PCL and (b) PCL/SrBG are after 1 day culture, and (c) PCL and (d) PCL/SrBG are after 7 days culture. FDA (green fluorophor), indicates live cells, while PI (red fluorophor) indicates dead cells. Scale bars are as shown.

Page 77: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 59

Figure 4.4 SEM images of MC3T3 cells cultured on PCL (a)(b) and (c) and PCL/SrBG (d)(e) and (f) scaffolds. Time points are: 3 days (a) and (d), 7 days (b) and (e), and 14 days (c) and (f). Scale bars are as shown.

Figure 4.5 Confocal laser scanning microscopy images of MC3T3 cells cultured on melt-electrospun PCL/SrBG (a) and PCL scaffolds (b) for 3 days. Green: Alexa Fluor 488 Phalloidin conjugates (actin), Blue: DAPI (nuclei), and Red: Alizarin red S (SrBG particles). Scale bars are as shown.

Page 78: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

60 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Figure 4.6 MTT metabolic activity assay of MC3T3 cells over 28 days culture (n=6). Error bars = ± SD of mean. * indicates significant increase in metabolic activity (p < 0.05).

ALP activity and mineralisation (Cell differentiation)

Normalized ALP activity (per cell) in all four experimental groups showed

increasing values with length of culture time and peaked on day 21 as presented in

Figure 4.7. The PCL/SrBG_O group showed significantly higher ALP activity when

compared to PCL_O group (p < 0.05) at both the 21 and 28 day time points (Figure

4.7). Scaffolds cultured in osteogenic media (PCL_O and PCL/SrBG_O groups)

showed significantly higher ALP activity compared to control groups (PCL_C and

PCL/SrBG_C groups) at all time points after 7 days. Alizarin red S staining indicated

more calcium mineralization (dark red) formed over the duration of cell culture and

the stain appeared darker in both PCL/SrBG groups compared to the PCL scaffolds

(Figure 4.8). Scaffolds cultured without cells (PCL Control and PCL/SrBG Control)

showed no staining and low levels of staining, respectively.

Page 79: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 61

Figure 4.7 Normalised ALP activity of MC3T3 cells cultured on melt-electrospun PCL and PCL/SrBG scaffolds cultured in osteogenic and control media (n=4). ALP activity is divided by a total number of cells based on the DNA content obtained via PicoGreen assay. Error bars = ± SD of mean. * indicates significantly higher ALP activity per cell in the indicated group compared to the PCL_Control group of the time point (p < 0.05), † indicates significantly higher ALP activity per cell compared to the PCL_Osteogenic media group of the time point (p < 0.05).

Page 80: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

62 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Figure 4.8 Alizarin red S staining of PCL and PCL/SrBG scaffolds cultured with MC3T3 cells in control and osteogenic media over 28 days. Control scaffolds are unseeded but have been cultured for the same periods of time in culture media.

Expression of specific genes by MC3T3 cells on both scaffolds

Normalized RT-qPCR results indicated peak expression of ALP gene on day 21

(Figure 4.9a) and OCN gene on day 14 (Figure 4.9b). PCL/SrBG_O scaffolds showed

significantly up-regulated ALP expression levels after 7 and 21 days compared to

PCL_O. There was no significant difference in ALP expression between control

groups (PCL_C and PCL/SrBG_C). The results also showed significantly higher OCN

expression level in the PCL/SrBG_O groups compared to PCL_O after 14 days in cell

culture.

Page 81: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 63

Figure 4.9 Gene expression of osteoblast markers ALP (a) and OCN (b), in all experimental groups over 28 days (n=9). Data represents mean + standard deviation. Error bars = ± SD of mean. * indicates a significant increase in gene expression compared to PCL_O (p < 0.05).

Extracellular matrix (ECM) formation

Van Gieson staining showed that the concentration of collagen increased with

length of time in culture in all scaffold groups (Figure 4.10). Among them, PCL_O

and PCL/SrBG_O showed higher levels of collagen formation compared to the PCL_C

and PCL/SrBG_C groups. Also, PCL/SrBG_C and PCL/SrBG_O groups showed

noticeably higher ECM formation compared to the PCL_C and PCL_O groups.

Page 82: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

64 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Figure 4.10 Van Gieson staining of PCL and PCL/SrBG scaffolds cultured with MC3T3 cells in control and osteogenic media over 28 days. Control scaffolds are unseeded but have been cultured for the same periods of time in culture media.

4.4 DISCUSSION

Organic polymer/inorganic particle composites have been fabricated into

scaffolds via the technique of solution-electrospinning [160–162] in the past, however,

the use of melt-electrospinning, which has prominent advantages over its solution

counterpart has gained little attention in the field of tissue engineering [83]. I was able

to successfully establish and optimise a technique for melt-electrospinning PCL/SrBG

composite fibre scaffolds, for the first time, which show promise for application in the

bone TE field. The complexity of melt-electrospinning PCL composite scaffolds

occurs due to the inclusion of the inorganic SrBG phase. Large clumps of SrBG can

interfere with the continuity of fibre production by blocking the nozzle of the spinneret,

causing the formation of beads, and an agglomeration of inorganic particles. A similar

situation also occurs when solution-electrospinning scaffolds comprising inorganic

and organic phases, and to address this problem in solution-electrospinning, a

surfactant can be introduced to ensure the homogeneous dispersion of the inorganic

Page 83: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 65

particle phase throughout the organic polymer [163]. However, this introduces another

impurity to the scaffold which can potentially contribute to increased toxicity to cells

(in addition to the organic solvent). In this study, I circumvented these issues by

manually dispersing the SrBG particles into molten PCL bulk until a homogeneous

distribution was achieved via mechanical mixing. The SrBG particles maintained an

even dispersion during the entire electrospinning process (as demonstrated with μCT

scanning showing an even distribution of particles in Figure 4.1f) this is due to the

molten PCL (at ~60 °C) being sufficiently viscous to prevent any detectable

sedimentation effect occurring. Using this novel approach of composite fabrication by

melt-electrospinning, we were able to reproducibly manufacture a scaffold with

microfibers and with pore sizes large enough to allow for cellular infiltration and

nutrient diffusion [164]. A loading of 10 wt% SrBG particles were selected in this

study based on previously published literature on polymer/BG composite materials

[14,165–167] and on ongoing studies in our team [14].

The parameters used to produce PCL/SrBG scaffolds via melt-electrospinning

technique were explored based on the ones to produce PCL scaffolds due to lack of

previous experience in TE field. I aimed to produce PCL/SrBG composite scaffolds

with smooth fibre jets of random deposition (as that of PCL) scaffold by altering key

parameters of melt-electrospinning shown in Table 4.1. Heating temperature during

the electrospinning process is one of the essential parameters, the PCL/SrBG

composite fibre started to break to show a ‘spitting’ phenomenon when the

electrospinning temperature is too high and the composite was too viscous to

electrospin and blocked the needle when it was too low. This process to produce

continuous composite fibres to form melt-electrospun scaffolds was defined as

optimisation in study 1.

Previous work has determined that the fibre diameter of the melt-electrospun

PCL scaffolds (5–33 μm) is optimal for supporting cell culture [90,164]. Due to the

slightly different conditions required for successful production of melt-electrospun

PLC-SrBG scaffolds compared to melt-electrospun PCL ones, the PCL/SrBG fibres

(46.1 +/- 16.6 µm) were slightly larger than the PCL ones (30.6 +/- 1.8 µm) produced,

as were the inter-fibre spacing within the scaffold. One of the reasons for this

difference owed to the addition of the SrBG particles which has an effect on the

thermal properties and crystallinity of the PCL component [147], requiring higher

Page 84: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

66 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

temperatures to obtain similar viscosity. The presence of occasional large SrBG

particles up to 38 microns in size created a few very large fibres in the PCL/SrBG

scaffolds, resulting in a greater variance in fibre diameter of PCL/SrBG than the PCL

alone. It is well established in electrospinning that fibre size is proportional to inter

fibre spacing, so it is to be expected that the slight increase in fibre size led to an

increase in inter fibre spacing of the scaffolds. However, both scaffold types had no

significant difference in measurable weight and had a similar gross morphology when

examined by light microscopy and µCT (Figure 4.1a and 4.1b, 4.1e and 4.1f).

The ability for PCL/SrBG scaffolds to promote osteogenesis is dependent upon

the SrBG particles leaching ions into the local environment; as previously discussed,

the Sr ion has been shown to increase osteogenesis by enhancing osteoblast metabolic

activity and ALP activity [58]. Strontium incorporated biomaterials have been

increasingly interesting as Sr ions have great affinity to bone tissue were found to be

able to stimulate bone formation and reducing bone resorption [168–170]. As a result,

Sr has been incorporated into bioactive ceramics including bioactive glass for

enhanced bioactivity [58,171–173].The SrBG with 75% Sr substitution used in this

study was provided by our collaborator – Prof. Molly Stevens’s group. At the

development stage of the SrBG, serial percentages (0-100%) of Sr substitution were

studied [57,58]. According to the results, even though the SrBG with 100% Sr

substitution showed best promotion of osteoblast activity and inhibition of osteoclast

activity of cells cultured on these materials compared to lower substitutions [58], the

high Sr content in the SrBG also inhibited osteoblast proliferation [57]. As a result, the

Sr75 SrBG was a balance between the efficacy and toxicity of Sr ions. I confirmed that

the PCL/SrBG composite scaffolds were releasing Si and Sr ions into the media,

therefore, the scaffolds offer potential to promote osteoblasts differentiation [58]. The

5g/L SrBG scaffold to media ratio was adopted from previous studies for easy

comparison [171,174]. The scaffolds were immersed in the media and the containers

were sealed to minimise the risk of bacterial and fungal contamination, and the media

were collected for analysis at predetermined time points. It was also observed that the

ion release rate was very slow for the initial three days, but then rapidly increased,

most likely due to surface hydration of the PCL fibres resulting in an increase in the

direct exposure of SrBG particles to the media. Additionally, the presence of SrBG

particles stimulated CaP deposition on the surface of scaffolds, detected by the

Page 85: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 67

decrease of Ca and P ions in the media, and confirmed directly by EDX (Figure 4.2a).

According to the literature, the exposed SrBG particles may initiate the surface

mineralization by providing nucleation sites [175]. This would also explain why a CaP

layer was not present on PCL scaffolds subjected to the same conditions. In the bone

TE field, CaP coating is a popular approach to enhance bioactivity of polymeric

scaffolds by decreasing their surface hydrophobicity and thus creating scaffolds which

can better mimic the natural structure of bone ECM [102,176–180]. As a mineralized

surface is known to be favourable for cell attachment and migration, the inclusion of

SrBG particles in PCL fibres has provided a mechanism for the PCL/SrBG scaffolds

to present a higher degree of bioactivity than PCL scaffolds alone.

As expected, both PCL and PCL/SrBG scaffolds were capable of facilitating cell

attachment and proliferation, as shown using MC3T3 cells. After 1 day and 7 days

culture, negligible numbers of dead cells could be found on either scaffold type (<

10%), and live cells were evenly distributed across the scaffold, suggesting that

attachment and migration were not significantly altered by the incorporation of SrBG

to the PCL component, or the increase in fibre diameter (Figure 4.3). The cell viability

(over 90%) is high when compared to previous studies [181,182] and suggests that the

fibres encourage cell growth and proliferation. Quantitative results obtained by the

MTT assay suggest that the addition of SrBG particles did not have any significant

effect on the proliferation of the cells upon the scaffolds, and visualising the cells via

SEM showed that the fibres were acting as structural support for cells, with strong

attachment, and bridging of the larger pores in both scaffolds (Figure 4.4). It is

expected that these interconnective pores provide a large volume of space for cells to

proliferate and infiltrate, and allow nutrient transfer within the structure of the scaffold.

While cells could attach and proliferate on both scaffolds, a distinct difference between

the attachment of cells onto PCL and PCL/SrBG scaffolds was detected via CLSM

(Figure 4.5). The presence of SrBG particles directed cellular attachment to be more

localised around the particles, and this observation is in line with the established

literature, confirming the formation of a strong bond between SrBG and living tissue

[148]. This again supports the theory that the organic fibre/inorganic particle system

better mimics the natural structure of bone than polymers alone and can facilitate cell

attachment, proliferation and formation of appropriate tissue.

Page 86: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

68 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

Higher mineralization was observed in the PCL/SrBG_O than the other three

groups as identified by the qualitative results of Alizarin red S staining. This suggests

that even though it has been established that SrBG has the capacity to enhance

mineralization of osteoblasts, this has not been directly observed without additional

osteogenic factors introduced into the media. It should be noted that there is

background staining present on the scaffolds with the inclusion of SrBG (control

scaffolds after 28 days of incubation in media) as the Alizarin red S dye binds to

calcium which is a component of the bioactive glass. However, by including negative

controls in our experiments (Figure 4.8) I illustrate that this staining level is

significantly lower than the groups with the cell inclusions, implying that the

mineralisation detected is more likely attributed to the cell response (mineralisation)

to the bioactive glass and/or osteogenic media. As shown in Figure 4.2, the precipitated

CaP layer could also contribute to the positive staining by Alizarin Red S due to the

nature of this stain. This explains why the PCL/SrBG scaffolds with cells showed

overall more intense staining compared to PCL control scaffolds. The effect of CaP

layer was more dominant at the early time point (day 7) when the cell ECM

mineralization level was low, which explains why some scaffolds in PCL/SrBG_C

group stained as intensely as the ones in PCL/SrBG_O group. In order to further

support the osteogenic effect of the SrBG component, the expression of two key genes

(ALP and OCN) which are expressed when osteoprogenitors cells commence

differentiation into osteoblasts [183,184], were investigated. Normalized ALP activity

confirmed up-regulated osteoblast differentiation on PCL/SrBG_O scaffolds

compared to PCL_O scaffolds alone, in the presence of osteogenic media after 21 days.

At earlier time points, the ALP activity per cell was not significantly different between

the two groups. However, after 3 weeks in culture, there was significantly higher ALP

activity in the PCL/SrBG_O group than in the PCL_O one. This increase of ALP

activity at 21 days is most likely induced by the presence of ions from the SrBG eluting

into the media (Si and Sr), as the ion dissolution studies show that relatively little

elution occurs for the first 3 days, followed by a more rapid release building up to 14

days, it is possible that this delayed release is responsible for postponing an observable

effect until 21 day culture. As Gentlemen et al. discussed previously[58], the increased

amount of Si and Sr ions released from PCL/SrBG scaffolds works synergistically to

better promote osteoblast differentiation compared to PCL scaffolds alone. The up-

regulation of the ALP gene, which we observed, correlates with the observed ALP

Page 87: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 69

activity. The significantly higher OCN gene expression present in PCL/SrBG scaffolds

compared to the PCL ones after 14 days culture further suggests an additional

osteogenic capability of the composite scaffold compared to PCL scaffolds. However,

I found no such significant difference between groups without osteoinductive reagents

(PCL_C and PCL/SrBG_C) indicating that the SrBG present in the composite scaffold

alone was not sufficiently osteogenic to induce differentiation alone. Our data does

suggest that the incorporation of SrBG particles into the PCL scaffold can enhance the

osteogenic potential of cells when paired with osteogenic media to a higher degree

than the osteogenic media alone. A possible reason for the lack of osteogenic response

from PCL/SrBG scaffolds alone could be an insufficient amount of SrBG particles

present within the scaffold (10 wt%). Commonly, cytokines (typically ascorbic acid,

β-glycerophosphate and dexamethasone) are used for osteoblasts differentiation in

vitro[185]. However, in the previous study by Gentleman et al., osteogenesis was

observed when the human osteosarcoma cell line Saos-2 cells were cultured on SrBG

discs in standard culture media (RPMI 1640 media supplemented with 10% (v/v) foetal

bovine serum (FBS) and 2 mM L-glutamine) without any osteogenic reagents such as

ascorbic acid [58]. Based on these results, we decided to explore the osteogenic

capacity of PCL/SrBG scaffold without any other osteogenic factors. In Gentleman’s

study, cell culture media were treated with a total of 1.5 mg/ml of SrBG powder for

ion dissolution and found significantly enhanced osteoblasts proliferation and

differentiation [58]. In our experiment, the average weight of a PCL/SrBG scaffold

was 2.6 mg, with only 10% of the weight being SrBG particles, so there was an average

SrBG concentration of 0.325 mg/ml cell culture media available for ion dissolution,

with not all of the particles exposed at one time. Unsurprisingly, Sr2+ and Si4+

concentration in the PCL/SrBG composite scaffolds dissolution media was much

lower than those observed by Gentleman et al. when they investigated SrBG particles

alone, based on elemental analysis. In addition to the lower total SrBG present, the

PCL component acts as a physical barrier or coating inhibiting the burst release of the

ions from the SrBG particles present within the core of the PCL fibres. However, our

system has the advantage of facilitating a longer term sustainable ion release (releasing

ions as the PCL fibres degrade thus exposing the SrBG), an effect which is more

desirable for bone regeneration compared to the burst release observed of SrBG alone.

However, in order to enhance osteogenic potential of this composite PLC/SrBG

scaffold sufficiently to elicit an effect in in vivo bone repair, it will most likely be

Page 88: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

70 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

necessary to increase the weight percentage of the SrBG component which would

involve higher loading during the melt-electrospinning process, which is a difficult

undertaking owing to complexities in continuous polymer fibre production when

higher concentrations of particles are present. One future approach would be to surface

coat the fibres with additional SrBG. Another approach to increase the rate of release

of the SrBG ions is to etch the PCL/SrBG scaffold for longer with NaOH, this is a

procedure which is already employed with PCL scaffolds in order to increase the

hydrophobicity of the scaffold, and the technique can be used to expose a higher

amount of SrBG to the media. I undertook surface etching treatment to allow for

maximum SrBG particle exposure and reached a critical time limit of 12 hours at which

time the etching was leading to particles “falling” out of the polymer matrix, therefore

it is important to have a trade-off between etching time and not losing important SrBG,

and also not to degrade the PCL matrix to an extent where the fibres may become

mechanically compromised, in this study I opted for one hour etching as I observed a

greater number of particles exposed at this time point with no particles falling out of

the PCL matrix.

In natural bone, collagen-based extracellular matrix (ECM) is essential for the

structural support of cells and mineral contents [6]. As such, I looked into the amount

of collagen deposited by cells onto the scaffolds in the experimental groups and found

that cells on the composite scaffolds appeared to produce a greater amount of ECM

than the PCL scaffolds alone (Figure 4.10). This was a qualitative observation which

suggests that SrBG may have other stimulatory effects, in addition to enhancing

mineralization.

In addition to electrospinning, there are many other techniques available in TE

to incorporate an inorganic phase such as SrBG to a polymeric scaffold. For example,

BG coating of polymers is another common approach [50] which is relatively easy and

quick method for the incorporation of BG particles onto the polymeric scaffolds

surface. In comparison to melt-electrospinning, this technique facilitates the maximum

contact of bioglass to cells initially possible; however, this approach can potentially

reduce the interconnective porosity of scaffolds by clogging the spaces between fibres

with bioglass particles. Additionally, the technique requires the use of organic solvents

involved in the coating process which can pose potential toxic effects to living cells

and tissues if they are not efficiently removed prior to cell exposure. It would be

Page 89: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration 71

desirable to produce a sustained release of ions from a bone TE scaffold, which cannot

be easily achieved from BG coating alone due to the burst release nature of ions from

BG particles which are highly exposed on the surface of the composite [186]. The

technique of melt-electrospinning enables an even distribution of BG particles

embedded (and exposed through etching) throughout the scaffold fibres which is

suitable for long-term sustainable ion release. Approaches to increase the percentage

of SrBG in the polymer bulk, combined with surface coating approaches should be

investigated to exploit the potential these highly porous melt-electrospun scaffolds

show in the field of bone TE.

4.5 CONCLUSIONS OF STUDY 1

An organic/inorganic composite biomaterial was produced for the first time by

incorporating SrBG particles into PCL bulk and melt-electrospinning to form highly

porous fibrous sheets. The composite scaffolds produced facilitated cellular

attachment and proliferation, and possessed enhanced osteogenic potential compared

to PCL scaffolds alone. ALP activity of MC3T3 cells cultured on these composite

scaffolds was enhanced and increased osteoblast differentiation was observed through

up-regulation of gene expression (ALP and OCN) in vitro. The composite scaffolds

also enhanced collagen deposition. These results showing enhanced osteogenesis

support similar studies using SrBG particles alone. The simple processibility of PCL

allows it to provide a potential platform for delivering the SrBG component and melt-

electrospinning has demonstrated promise as a fabrication technique which can

produce PCL/SrBG scaffolds which are highly porous, and these results show potential

for the technique to be further developed for the application of designing a versatile

biomaterial composite for the bone tissue engineering arena.

The 10 wt% PCL/SrBG scaffolds showed enhanced osteogenic capacity in

osteogenic media. However, their practical application is limited mainly due to the

lack of osteoinductivity, which means the composite scaffolds are not yet the ideal

solution for bone defect repair. In addition, another limitation is that the structure of

the scaffolds produced using melt-electrospinning in this study presented in a random

mesh layout instead of controlled size and shapes. Therefore in the subsequent study

2, I planned to further enhance the bioactivity of the PCL/SrBG scaffolds by increasing

the proportion of SrBG contents in the composite material and optimise the melt-

Page 90: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

72 Chapter 4: (Study 1) Fabrication and In vitro investigation of PCL, 10 wt% PCL/SrBG electrospun scaffolds for bone regeneration

electrospinning process to produce PCL/SrBG composite scaffolds with enhanced

bioactivity (ideally making the scaffolds osteoinductive) and controlled fibre

deposition.

Page 91: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 73

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

In study 1, I produced 10 wt% PCL/SrBG scaffolds which showed enhanced

osteogenic capacity compared to PCL only scaffolds but the results indicated that there

was not enough SrBG filler phase to make the composite with better osteogenic

properties. In this study, in order to further enhance the bioactivity of composite

scaffolds, I increased the weight percentage of SrBG particles in the PCL bulk. To

minimize the risk of needle blockage and obtain a homogeneous composite, I reduced

the SrBG particle size by milling and improved PCL/SrBG composite preparation

techniques. I then optimised the melt-electrospinning technique to produce PCL/SrBG

composite scaffolds with higher SrBG contents in a direct writing mode to obtain

scaffolds with ordered layer-by-layer fibre deposition. These new PCL/SrBG scaffolds

with increased SrBG, which was up to 50 wt% of PCL, were investigated in the same

manner as the scaffolds in Chapter 4 with an in vitro cell study for their osteogenic

properties. The results of this study of PCL/SrBG (50 wt%) scaffolds can be used to

determine whether these improved scaffolds are a potential patient-specific solution

for bone defect healing.

5.1 INTRODUCTION

Bone has a remarkable self-healing capacity to repair itself scarlessly after injury

[24]. However, this capacity can be impeded when the large area of bone loss exceeds

its regenerative capacity [26]. While the classic bone-grafting clinical treatment for

large bone defects renders suboptimal results, the scaffold-based tissue engineering

approach offers an off-the-shelf alternative [26]. Recently a paradigm shift has

occurred towards tissue engineering smart biomaterials with patient specific structures

[12,44,166]. These ‘intelligent’ bone scaffolds are expected to function as a temporary

Page 92: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

74 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

matrix to allow for cell recruitment from nearby host tissues, cell proliferation and cell

differentiation towards osseous tissue, while maintaining the mechanical support to

the defect site [23]. In spite of the recent advance in biomaterials and fabrication

techniques in TE field, it is still challenging to produce fully functioning scaffolds for

bone regeneration.

Materials selection and structural design are two key research focuses towards

the development of ideal scaffolds for bone. Synthetic materials including polymers

and bioactive glass (BG) are common for bone scaffold fabrication [50,70]. Among

the synthetic polymers, polycaprolactone (PCL), which is an FDA-approved aliphatic

polyester, has become a promising candidate of bone scaffold material due to its

favourable rheological and viscoelastic properties [45]. Bioactive glasses (BGs) are a

sub-category of bioactive ceramics that bond with host tissues via the formation of a

biologically active hydroxyl carbonate apatite (HCA) layer on the surface [16]. The

ions released from BGs make them superior bone forming materials to other

bioceramics by stimulating osteogenic differentiation [16,50]. Since Larry Hench

developed the first BG - Bioglass®, known as 45S5, a variety of BGs have been

introduced over the years [16]. Strontium-substituted BG is one of the derivatives that

modified the 45S5 BG by substituting 0-100% of the calcium component of the 45S5

formulation with strontium [57], and it has shown superior osteogenic capacity to 45S5

BG [58].

In isolation, polymer alone or bioactive glass alone is considered suboptimal for

bone defect healing. However the polymer/BG composite materials, which exploit the

processability of polymers and bioactivity of BG filler phase, have been introduced to

TE as a new family of bioactive materials [50]. Previously, polymer/BG composite

scaffolds have been developed for TE purposes. However, these scaffolds were mainly

produced with traditional manufacturing techniques (thermally induced phase

separation [187], solvent casting [188], microsphere sintering [68]), which are limited

to produce scaffolds with simple geometries and uncontrolled internal architectures.

Alternatively, melt based additive manufacturing techniques have been employed to

produce scaffolds with desired geometry and controlled internal architecture [14,189].

Among these techniques, melt-electrospinning is a versatile fibre-based technique for

scaffold production with smaller fibre diameter and thus larger surface area to volume

ratio compared to melt extrusion. Recent developments the direct writing melt-

Page 93: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 75

electrospinning techniques make use of the stable and predictable fibre deposition to

fabricate scaffolds in a layer-by-layer manner [93,94]. However, the range of candidate

materials that can be used for melt-electrospinning is restricted by their viscoelastic

properties [190] and therefore melt-electrospun polymer/BG composite scaffolds are

rarely reported. In order to minimise this limitation, hybrid electrospinning systems,

which combine the processes of individual solution- and melt-electrospinning, have

been proposed but with limited application [86,97,98].

In this study, I introduced a novel solution-assisted hybrid melt-electrospinning

system for the production of PCL/SrBG composite scaffolds with increased SrBG

filler phase (50% in PCL) compared to our previous study (chapter 4) [191]. I report

for the first time, obtaining continuous and stable composite fibres producing 50%

PCL/SrBG composite scaffolds in a layer-by-layer manner with 0/90° cross-hatched

deposition. I furthermore characterised the scaffolds and assessed their bioactivity in

vitro. The in vitro study results indicated that the PCL/SrBG scaffolds showed

enhanced osteogenic capacity compared to PCL ones.

5.2 MATERIALS AND METHODS

5.2.1 PCL/SrBG composite preparation

SrBG with the composition of 46.13 SiO2 – 2.60 P2O5 – 24.35 Na2O – 26.91

(SrO:CaO) (mole %) where 75% of the calcium was substituted with strontium was

prepared as previously described [57,191]. SrBG frits were ground and sieved to yield

particles 20 μm to 100 μm in diameter and then provided to us by our collaborator

Professor Molly Stevens’ group at Imperial College London, UK.

Particle size optimisation

In order to obtain homogeneous distribution, reduce the risk of needle blockage

and to increase the surface area of SrBG particles, the SrBG particles were ground with

a micronizing mill (McCrone Microscopes & Accessories, USA). Briefly, 2.5g of

SrBG particles were loaded into a plastic chamber filled with zirconia beads and then

wetted with 13ml of absolute ethanol. A series of incremental grinding times (from

30s to 8h) were used to obtain the optimal grinding time, particle sizing was performed

at each time point with a Malvern Mastersizer 3000 (Malvern, UK). The particles were

traditionally dried in the oven at 60 °C but aggregation was seen to occur indicated by

increased particle size. To minimise particle aggregation, I used freeze drying instead

Page 94: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

76 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

of oven drying, to ensure particle size distribution remained in the same range as

ground.

PCL/SrBG composite preparation

The ground SrBG particles were incorporated into the PCL bulk by fast

precipitation into excess ethanol [14,192]. Briefly, 10% (w/v) PCL solution was

prepared by dissolving 2g of PCL pellets (CAPA 6500, Perstorp, UK) into 20 ml of

chloroform (MERCK, Millipore, Australia) at room temperature. Next, 1g of SrBG

particles were added into PCL solution for a composite of 50 wt% PCL/SrBG. The

composite solution was homogenised by constant stirring for 8 h with 2 x 2min

ultrasonication in between. The homogeneous mixture was then precipitated into a 10-

fold excess of 100% ethanol (MERCK, Millipore, Australia). The 50 wt% PCL/SrBG

composite was then collected and air dried in the fumehood to evaporate the solvents.

5.2.2 Scaffold fabrication

Both PCL/SrBG composite and PCL control scaffolds were fabricated with the

electrospinning rig built in house at QUT. The outstanding features of this rig include

positive/negative dual power packs and motorised stage that allows layer-by-layer

fibre deposition [193]. PCL/SrBG composite scaffolds were fabricated using a novel

hybrid melt-electrospinning technique in the direct write mode. PCL control scaffolds

were fabricated using direct writing melt-electrospinning (link to video:

https://www.youtube.com/watch?v=EzhMCzn8C80). Briefly, every 1g of composite

PCL/SrBG composite was mixed into 1 mL of chloroform in a 2 mL syringe. The

syringe was then inserted into a water jacket installed in the rig and the temperature

was maintained at 60 °C by a water bath. The PCL scaffolds were melt-electrospun at

80 °C. All scaffolds were fabricated with a 90° cross-hatched laydown pattern and

fibre spacing of 1mm (links to videos: https://www.youtube.com/watch?v=5ZB-

PCpw8qU and https://www.youtube.com/watch?v=nlRtngXuoJE). The optimised

electrospinning parameters are summarised in Table 5.1.

Table 5.1 Parameters for 50 wt% PCL/SrBG and PCL scaffolds fabrication Scaffold type

Voltage Temperature

Collection distance

Needle gauge

Flow rate

Feed speed

PCL 3.5/-3.2 KV 80 °C 1 cm G20 45 μl/h 800 mm/min PCL/SrBG 3.5/-3.2 KV 60 °C 0.5 cm G20 95 μl/h 800 mm/min

Page 95: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 77

5.2.3 Scaffold characterisation

Microscopy

A light microscope with ZEN blue software (Zeiss Axio Imager M2, Germany)

and a Sigma field emission scanning electron microscope (FESEM, Zeiss, Germany)

were used to examine scaffold surface morphology and fibre diameter.

Confocal microscopy

PCL/SrBG and PCL scaffolds were stained for 5 min with Alizarin red S (Sigma,

Australia) dye solution (1 g of powder into 50 ml of distilled water) 150 µl was added

to each scaffold. The scaffolds were washed repeatedly with MiliQ water until the

solution ran clear. The scaffolds were imaged with a Nikon A1R confocal microscope

(Nikon, Australia).

Backscattered SEM imaging

The PCL/SrBG scaffolds were embedded in epoxy resin and ground down until

the scaffolds fibres were exposed to the resin surface. The embedded scaffolds were

imaged with SEM (JEOL JXA 8530F Hyperprobe, USA) to detect backscattered

electron (BSE) for the qualitative analysis of the elemental composition on the surface

of the PCL/SrBG scaffolds.

Ion dissolution and precipitation

PCL/SrBG and PCL scaffolds were incubated at 37 °C/5% CO2 in alpha-

Minimum Essential Medium (α-MEM, Invitrogen, Australia) supplemented with 1%

(v/v) of penicillin-streptomycin (Invitrogen, Australia) at w/v of 5g/L. The media were

collected at 3, 6 hours, 1, 2, 3, 7, 14, 21 and 28 days for further elemental analysis.

The ion concentration of calcium (Ca2+), phosphate (PO43-), silicon (Si4+) and

strontium (Sr2+) in α-MEM was assessed with an Agilent 8800 Inductively Coupled

Plasma Mass Spectrometer (ICP-MS, Agilent, USA). The scaffolds retrieved from the

media were air-dried under vacuum for 48 hours and scanned with energy-dispersive

X-ray spectroscopy (EDX) (Sigma FESEM, Zeiss, Germany) to identify elemental

deposition on the surface of fibres.

5.2.4 In vitro studies

To examine the osteogenic potential of the PCL/SrBG composite scaffolds with

50 wt% SrBG loading and to compare their bioactivity to the scaffolds produced in

study 1, in vitro cell culture experiments similar to were carried out using mouse

Page 96: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

78 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

osteoblast precursor cell line, MC3T3-E1 (passage 9). A series of in vitro assays were

used to assess cell viability and metabolic activity, proliferation, osteoblast

differentiation and gene expression.

Cell culture

Prior to experimentation, MC3T3 cells (sub-clone 14) [152] were cultured for

expansion in growth media: α-MEM cell culture media supplemented with 10% (v/v)

foetal bovine serum (FBS, Invitrogen, Australia) and 1% (v/v) penicillin-streptomycin

at 37 °C/5% CO2. Scaffolds (both types) were etched in 5M NaOH for 15min at 37 °C

to enhance initial cell attachment and cut with a 6 mm biopsy punch. The scaffolds

were sterilised by 30min immersion in 70% ethanol and air dried in the fume hood,

followed by UV irradiation for 20 min each side. The PCL/SrBG (50 wt%) scaffolds

were conditioned in α-MEM supplemented with 1% (v/v) penicillin-streptomycin 1

week prior to cell seeding. MC3T3 cells were seeded at a density of 50,000 cells per

scaffold, and after 24 hours culture, half the samples were treated with osteogenic

media (growth media supplemented with 10mM β-glycerophosphate, 50 µg/ml

ascorbic acid and 0.1mM dexamethasone (all osteogenic supplements were supplied

by Sigma)), the other half were cultured in growth media as a control. A total of four

different groups were studied as shown in Table 5.2 for the subsequent experiments.

Table 5.2 In vitro experimental groups of study 2. PCL/SrBG scaffolds (50 wt%) were studied in both growth media and osteogenic media and PCL scaffolds were studied as control.

Group Scaffold type Culture media type

n

1. PCL_C PCL Growth (control) 125

2. PCL_O PCL Osteogenic 125

3. PCL/SrBG_C PCL/SrBG Growth (control) 125

4. PCL/SrBG_O PCL/SrBG Osteogenic 125

LIVE/DEAD staining

To determine both the cell viability on the scaffolds and their distribution,

LIVE/DEAD assay was carried out as described in study 1. Briefly, after days 1 and

7, cell culture media were removed and the scaffolds were moved into a fresh plate,

and the scaffolds were washed twice with PBS. The scaffolds were then incubated in

solution containing 0.67 µg/ml fluorescein diacetate (FDA, Invitrogen) and 5 µg/ml

propidium iodide (PI, Invitrogen) for 5 min in the dark. The stained samples were

Page 97: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 79

visualized using a Zeiss Axio M2 Imager (Zeiss, Germany) fluorescent microscope (at

λ = 488 nm and λ = 568 nm excitation).

Assessment of cell attachment, morphology, and BG identification

Scanning Electron Microscopy (SEM) was used to examine cellular

attachment and morphology on the respective scaffolds. After 3, 7, 14, 21 and 28 days

in cell culture, the scaffolds were fixed and processed for imaging as described in study

1. Briefly, scaffolds were fixed in 3% glutaraldehyde immediately following cell

culture, and these scaffolds were washed in sodium cacodylate buffer (Sigma,

Australia) and 1% osmium tetroxides in cacodylate. After 2 washes in ultrapure UHQ

water (Millipore Australia, Australia), the scaffolds were dehydrated in ascending

grades of ethanol followed by Hexamethyldisilazane drying (all reagents were

supplied by ProSciTech, Australia). The sputter gold-coated scaffolds were visualized

using a Sigma field emission scanning electron microscope (FESEM, Zeiss,

Germany).

Confocal Laser Scanning Microscopy (CLSM) [155] was used to visualise the

morphology of actin fibres in green and nuclei of MC3T3 cells in blue on the scaffolds

and their interaction with the SrBG particles stained red by Alizarin red (Sigma,

Australia). As described in study 1, after 3, 7, 14, 21, 28 days, cell culture media were

removed and scaffolds were transferred into a fresh 48-well plate. Scaffolds were fixed

with 4% paraformaldehyde (PFA, Sigma) solution for 30 min at room temperature

after 2 careful washes with PBS. Samples were then washed with PBS and

permeabilized with 0.2% (v/v) Triton X-100/PBS solution for 5 min. Followed by 2

washes with PBS, samples were then incubated with 0.5% (w/v) Bovine serum

albumin (BSA, Sigma)/PBS for 10 min. The samples were stained by 0.8 U/ml Alexa

Fluor® 488 Phalloidin (Invitrogen) and 5 µg/ml DAPI in 0.5% (v/v) BSA/PBS

solution. After 1 wash with MiliQ water, the scaffolds were stained with Alizarin red

S (pH 4.2) for 5 min and washed with MiliQ water twice to remove excess stain. The

scaffolds were then stored in PBS until imaging. The PCL/SrBG scaffolds were

visualized with Leica SP5 Confocal microscope (Leica, Germany), images of

identically treated PCL only scaffolds were taken as controls.

Page 98: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

80 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Cell metabolic assay

MTT (3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; Sigma,

Australia) assay, a cell metabolic activity assay, measures the absorbance of formazan,

reduced from MTT by mitochondria in active cells [58]. The absorbance value reflects

the metabolic activity of the cells when cultured on the scaffolds in the four experiment

groups. Briefly, on days 1, 7, 14, 21 and 28, scaffolds (n = 4) were transferred into a

fresh 48-well plate containing 500 µl of fresh media supplemented with 20 µl of MTT

solution (5 mg/ml) each well. Scaffolds were incubated (37 °C/5% CO2) for 4 hours

in the MTT supplemented media, after which the media was removed and 100 µl (D1,

7), 200 µl (D14), 400 µl (D21) or 500 µl (D28) dimethyl sulfoxide (DMSO, Merck,

Australia) was added to each well. The plates were then covered with tinfoil and placed

on an orbital shaker for 10 min. After mixing, 100 µl of DMSO eluant from each well

was transferred into fresh 96-well plates and absorption at λ = 540 nm was measured.

The obtained reading was multiplied with the dilution factors of DMSO at all time

points respectively.

Cell osteoblastic differentiation and DNA quantification assays

In order to investigate the osteogenic potential of the PCL/SrBG scaffolds and

compare the osteoblastic differentiation of cells cultured on the composite scaffolds

and on PCL scaffolds, I measured Alkaline Phosphatase (ALP) activities of the cells

adhere scaffolds in 4 experimental groups at predetermined time points. ALP protein

was selected as it is a known marker of osteoblast differentiation and plays a key role

in mineralisation [58]. The ALP assay was coupled with a PicoGreen assay which

quantifies the DNA concentration. The DNA concentration can then be used to

calculate the cell number. Therefore I normalized the ALP activity by the cell number

to find the actual potential of osteoblastic differentiation of the MC3T3 cells.

For both assays, at the 7, 14, 21 and 28 day time points, cell culture media was

removed and scaffolds were washed twice with PBS and transferred into sterile 1.5ml

Eppendorf tubes containing 500 μl of 0.2 Triton-X/ ×1 TE buffer each. The collected

samples were stored at -80 °C until further processing. At the day of assays, the tubes

containing scaffolds were vortex for 6 × 30s each to lyse the cells off the scaffolds,

and the cell lysates were transferred into fresh 1.5 ml Eppendorf tubes. From this point,

Page 99: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 81

lysates were assayed for DNA content and ALP activity as described in section 4.2.3

of study 1.

Real-time quantitative polymerase chain reaction (RT-qPCR)

In order to further investigate the regulation of genes related to osteogenic

differentiation by the incorporation of SrBG component, the RT-qPCR was used to

quantify the relative expression of specific genes associated with early stage of

osteogenesis [159].

After days 7, 14, 21 and 28, cell culture media was removed and scaffolds (n =

9) were transferred into fresh sterile 1.5ml Eppendorf tubes containing 700μl Trizol

Reagent® (Invitrogen) with 3 scaffolds per tube. RNA was isolated according to the

manufacturer’s instructions.

The total quantity and purity of the extracted RNA were tested with a Nanodrop

Microvolume UV-Vis spectrophotometer (ThermoFisher Scientific, Australia). The

concentration of RNA was calculated to obtain a total of 500 ng cDNA per sample.

The reverse transcription process was carried out with DyNAmoTM cDNA Synthesis

Kit (Thermo Scientific, Australia) following manufacturer’s instructions.

RT-qPCR was performed on a 7500 Fast Real-Time PCR Systems (Applied

Biosystems ®, Australia) using SYBR Green as detection reagent following the same

procedure that has been reported previously [159]. The relative mRNA expressions of

ALP and Osteopontin (OPN) were assayed and normalized against the house keeping

gene β-ACTIN. Each sample was analysed in triplicate. The mean cycle threshold (Ct)

of each target gene was normalized against Ct of β-ACTIN; the relative expression

calculated using the following formula: 2-(normalized average Cts) ×104.

5.2.5 Statistical analyses

Statistical analysis was performed on results of all 4 groups at all time points

generated by MTT, ALP and RT-qPCR assays using two-way ANOVA with a post-

hoc Tukey test using IBM SPSS Statistics Software (Version 19). A p value of less

than 0.05 was considered statistically significant.

Page 100: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

82 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

5.3 RESULTS

5.3.1 Particle grinding and sizing

By comparing the SrBG particle size obtained from incremental series of

grinding times, 8 hours of grinding with the micronizing mill was determined to be the

optimal grinding time, where 90% of SrBG particles by volume (Dv90) were reduced

from 109 μm to < 6 μm. (The full experimental data can be found in Appendix B) The

freeze drying took place at -20 °C for 3 days and the dried particles did not show

significant aggregation of particles as shown in Figure 5.1.

Figure 5.1 SrBG particle size distribution before grinding, after grinding and after drying. The SrBG particles were ground by micronizing mill and size distribution and their size distribution was detected by Malvern Mastersizer 3000. As shown in the graph, the peak of particle size (green line) reduced from around 100 μm down to less than 1 μm after grinding (blue line). With the optimised drying method, the dried particles showed minimum agglomeration (red line).

5.3.2 Scaffold fabrication

After a series of optimization, the electrospinning parameters used for the

composite scaffold fabrication are shown in Table 5.1. PCL and PCL/SrBG composite

were successfully electrospun into 30×30×1.5 mm fibrous scaffolds, and both type of

0

2

4

6

8

10

12

14

0.01 0.1 1 10 100 1000 10000

Volu

me

Den

sity

(%)

Particle size (μm)

SrBG 8 hrs grinding

SrBG 8 hrs grinding-freeze driedSrBG before grinding

Page 101: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 83

scaffolds were cut into cylinders of 6 mm in diameter with a biopsy punch for further

analysis.

5.3.3 Characterisation

Scaffold morphology

Both PCL and PCL/SrBG scaffolds had similar morphology and showed 0/90°

cross-hatched laydown pattern under SEM. The PCL/SrBG scaffold showed a rough

surface with ‘brush’ like structures (Figure 5.2a, 5.2b, 5.2c, and 5.2d), while PCL

scaffolds showed smooth surface (Figure 5.2e, 5.2f, 5.2g, and 5.2h). The PCL/SrBG

fibres (75±21 μm) were larger in diameter to PCL ones (28±4 μm).

Page 102: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

84 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.2 SEM images of 50% PCL/SrBG (a-d) and PCL (e-h) scaffolds. Both PCL and PCL/SrBG scaffolds showed 0/90° cross-hatched deposition of electrospun fibres. With the zoomed-in images shown in c and d, surface of PCL/SrBG scaffolds were rough with mini fibre-like objects possible produced during scaffold fabrication. In comparison, g and h showed a smooth surface of PCL scaffolds.

PCL

/SrB

G

PCL

Page 103: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 85

SrBG particle distribution

As shown in Figure 5.3, the SrBG particles were stained in red by Alizarin red

as described in section 5.2.3 under a confocal microscope. These particles were found

all across the PCL/SrBG scaffold fibres. PCL scaffolds stained by the same technique

are not shown here as no fluorescence was detected by the confocal microscope. The

distribution was further assessed by a backscattered SEM, (Figure 5.4), Figure 5.4a

was an overview of a cross section of PCL/SrBG scaffold, and it showed a

homogeneous distribution of SrBG particles (bright spots) across the PCL fibres (dark

grey regions) in the whole scaffold. The elemental composition of the bright particles

were confirmed by the Sr elemental mapping, which indicated the areas with bright

spots matched the Sr rich areas as shown in Figure 5.4c and d.

Figure 5.3 Confocal laser scanning microscopy images of PCL/SrBG scaffolds stained by Alizarin red S. Red colour: SrBG particles. The composite scaffold fibres fluorescence red as Alizarin red bides to calcium in the SrBG components. The PCL scaffolds was stained with the identical conditions and as there were not biding sites for Alizarin red, the PCL fibres remained invisible under confocal microscope. Thus the images of stained PCL scaffolds were not included here.

Page 104: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

86 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.4 Backscattered SEM image of a PCL/SrBG scaffold. (a) an overview of the scaffold and inset (b) shows a zoomed in the central region of the scaffold. The bright spots are mineral contents and in this case the SrBG particles, whereas the grey areas indicate the contrast PCL material (b), and a zoomed-in image of the arrow-point area (c). To confirm this, a Sr element mapping was performed and (d) shows the distribution of Sr in the scaffold in green, which matches the area of bright spots. Overall, the distribution of SrBG components indicate a high loading and homogeneous distribution of the inorganic filler phase with the PCL fibres.

Ion dissolution

The elemental concentrations of Ca2+, PO43-, Si4+ and Sr2+ released from both

PCL and PCL/SrBG scaffolds into α-MEM media are shown in Figure 5.5(a–d). In the

media exposed to PCL/SrBG scaffolds, the Ca2+ and concentration (in the media)

decreased rapidly after 3 hours in cell culture, and then further decreased over the next

4 weeks. PO43- concentration of the PCL/SrBG scaffold dissolution media also

decreased over the period of 4 weeks. No significant change in Ca2+ and PO43-

concentrations was observed in cell culture media exposed to PCL scaffolds. In the

PCL/SrBG scaffold dissolution media, the concentration of Si4+ showed a sharp

increase after 3 hours and 6 hours in the media and became stable after 14 days in cell

culture. The Sr2+ concentration increased slowly in the first 2 days and increased

rapidly after 3 days and 14 days in the media. No change in Si4+ and Sr2+ concentrations

were observed in PCL scaffold dissolution media.

Page 105: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 87

Figure 5.5 Elemental concentrations of Ca2+ (a), PO4

3- (b), Si4+ (c), and Sr2+ (d) of a-MEM media incubated with 50% PCL/SrBG and PCL scaffolds over 28 days as determined by ICP-MS testing. The blue lines show the elemental concentration of PCL scaffolds dissolution media, whereas the red lines show the concentration of elements in PCL/SrBG scaffolds dissolution media. Overall, the Ca and P elements showed decreased concentration over time in the dissolution media of PCL/SrBG scaffolds and it remained constant in PCL scaffolds dissolution media (a and b). The Sr and Si elements had increased concentration in the dissolution media of PCL/SrBG scaffolds, and showed no increase in the PCL control samples (c and d).

CaP layer precipitation

The EDX analysis detected the presence of Ca and P elements on the surface of

PCL/SrBG fibres after 3 hours of scaffold immersion in α-MEM, which indicates the

formation of a CaP layer (Figure 5.6a). The CaP precipitation was not detected on PCL

surface and example EDX scanning results after 7 and 14 days in α-MEM shown in

Figure 5.6e and f respectively.

Page 106: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

88 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.6 Example surface elemental compositions of 50% PCL/SrBG scaffolds after 3h (a), 6h (b), 1 day (c) and 2 days (d) in a-MEM media as determined by EDX. The increase of Ca/Sr and P/Sr ratio indicated the continuous formation of CaP layer at composite scaffold surface. The results of PCL scaffolds were shown as control after 7 days (e) and 14 days (f).

PCL

/SrB

G

PCL

Page 107: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 89

5.3.4 In vitro studies

Attachment and proliferation of MC3T3 cells on scaffolds

The LIVE/DEAD staining showed viable MC3T3 cells (as shown in green)

evenly distributed across both PCL and PCL/SrBG scaffolds after 1 day cell culture

with negligible red stained cells detected. After 7 days, the cell number had

significantly increased (Figure 5.7). High magnification SEM images showed initial

attachment of cells on to both types of scaffolds (Figure 5.8a and c), and Figure 5.8b

and d showed a large number of cells covering scaffolds fibres and bridging to adjacent

fibres on scaffolds in all four groups. CLSM images showed numerous filopodia of

MC3T3 cells attaching onto the SrBG particles exposed on PCL/SrBG fibre surfaces

(Figure 5.9a) whereas cells attached and spread evenly on PCL fibres (Figure 5.9b).

The increase of MTT absorbance quantitatively indicated that PCL/SrBG composite

scaffolds were not cytotoxic by showing that the cell metabolic activity increased with

length of cell culture time in all four experimental groups (Figure 5.10). MTT results

showed a trend of cell metabolic activity in all experimental groups except PCL_O, in

which the activity peaked on day 21.

Page 108: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

90 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.7 LIVE/DEAD staining of MC3T3 cells cultured on melt-electrospun scaffolds. (a) PCL and (b) PCL/SrBG are after 1 day culture, and (c) PCL and (d) PCL/SrBG are after 7 days culture. FDA (green fluorophor), indicates live cells, while PI (red fluorophor) indicates dead cells.

Page 109: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 91

Figure 5.8 SEM images of MC3T3 cells cultured on PCL (a) and (b) and PCL/SrBG (c) and (d) scaffolds. Time points are: 3 days (a) and (c) (MC3T3 cells indicated by the yellow arrows), and 7 days (b) and (d) (MC3T3 cell sheets indicated by yellow arrows).

Figure 5.9 Confocal laser scanning microscopy images of MC3T3 cells cultured on melt-electrospun PCL/SrBG (a) and PCL scaffolds (b) for 3 days. Green: Alexa Fluor 488 Phalloidin conjugates (actin), Blue: DAPI (nuclei), and Red: Alizarin red S (SrBG particles).

Page 110: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

92 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.10 MTT metabolic activity assay of MC3T3 cells over 28 days culture (n=4). Error bars = ± SD of mean. * indicates significant increase in metabolic activity (p < 0.05).

ALP activity and mineralisation (Cell differentiation)

Normalized ALP activity (per cell) in all four experimental groups showed that

scaffolds cultured in osteogenic media (PCL_O and PCL/SrBG_O groups) showed an

overall higher ALP activity compared to control groups without osteogenic media

(PCL_C and PCL/SrBG_C groups) (Figure 5.11). More importantly, cells cultured on

PCL/SrBG scaffolds showed higher ALP activity compared to those on PCL ones

regardless of culture media type. At each time point, there is a clear trend of ALP

activity increase: PCL_C < PCL/SrBG_C < PCL_O < PCL/SrBG_O groups, except

day 21 where the PCL_O group showed lower ALP activity than PCL/SrBG_C group.

After 21 days of cell culture, the cells on PCL/SrBG scaffolds showed significantly

higher ALP activity when compared to PCL scaffolds in control media (p < 0.05).

After 28 days of cell culture, the PCL/SrBG_O group showed significantly higher ALP

activity when compared to PCL_O group and PCL/SrBG_C group (p < 0.05) (Figure

5.11).

Page 111: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 93

Figure 5.11 Normalised ALP activity of MC3T3 cells cultured on melt-electrospun PCL and PCL/SrBG scaffolds cultured in osteogenic and control media (n=4). ALP activity is divided by a total number of cells based on the DNA content obtained via PicoGreen assay. Error bars = SD of mean. * indicates significant difference (p < 0.05) between PCL/SrBG_C and PCL_C group at day 21 and PCL/SrBG_O and PCL_O group at day 28. ** indicates significant difference (p < 0.05) between PCL/SrBG_O and PCL/SrBG_C at day 28.

Expression of specific genes by MC3T3 cells on both scaffolds

The mRNA expression of ALP and OPN (osteogenic genes) were obtained by

RT-qPCR technique. The calculated relative expression of both genes in all 4 groups

at all time points were normalized by the ALP and OPN gene relative expression of

PCL_C group on day 7 to obtain the fold change of both genes respectively. Overall,

the fold change of ALP gene of PCL/SrBG_O group was higher than the other groups

over the cell culture period (Figure 5.12a). At day 21 time point, the ALP mRNA

expressed by cells in the PCL/SrBG_C group was significantly higher than the PCL_C

group; the ALP mRNA expression of PCL/SrBG_O group was significantly higher

than the PCL_O group and the PCL/SrBG_C group (Figure 5.12a). At 28 time points,

ALP mRNA expression of cells in the PCL/SrBG_C group was higher than the PCL_C

group. The RT-PCR results also showed significantly higher OPN expression level in

the PCL/SrBG_O group compared to PCL_O and PCL_C groups after 28 days in cell

culture (Figure 5.12b).

Page 112: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

94 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

Figure 5.12 Gene expression of osteoblast markers ALP (a) and OPN (b) as fold change to PCL control group in all experimental groups over 28 days (n=9). Data represents mean + standard deviation. Error bars = SD of mean. (a) * indicates significant difference (p < 0.05) of ALP mRNA expression between PCL/SrBG_C and PCL_C group at day 21 and PCL/SrBG_O and PCL_O group at day 21. (b) * indicates significant difference (p < 0.05) of OPN mRNA expression between PCL/SrBG_O and PCL_O group at day 28. ** indicates significant difference (p < 0.05) of OPN mRNA expression between PCL/SrBG_O and PCL_C at day 28 and (p < 0.05).

5.4 DISCUSSION

A number of composite materials of bioactive glass/biodegradable polymer

combination have emerged in the field of tissue engineering [194]. This new family of

bioactive materials presented in this thesis, which exploits the processability of

polymers with the stiffness, strength and bioactive character of the bioactive glass

fillers, are being increasingly applied, ranging from structural implants to TE scaffolds,

instead of the traditional single-material constructs [50]. Additive manufacturing

(AM) techniques are taking the place of traditional methods in 3D composite scaffold

fabrication as the AM scaffolds have the advantages of high porosity, interconnectivity

and tailorable size and shape [12]. Electrospinning was not technically considered as

an AM technique until the recent emergence of melt-electrospinning in a direct-writing

mode, which uses the same principle as that of fused deposition modelling (FDM) to

produce 3D scaffolds with much smaller fibre size [93,94]. Several polymeric

materials have been successfully produced into scaffolds via melt-electrospinning

technique. However, melt-electrospinning of polymer/inorganic particle composite is

rarely reported (As summarized in Table 5.3, a Web of Science search for ‘melt-

electrospinning’ AND ‘polymer composite’ generated 10 results on 10/10/2016 and

Page 113: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 95

only 2 of them as indicated by * were studies of polymer/inorganic composite

materials). This is mainly due to the fact the composite scaffold production via melt-

electrospinning has been proven an extremely difficult technique. Several factors

contribute to this situation. Firstly, the incorporation of inorganic filler phase greatly

changes the viscoelastic properties of polymers, which disturbs fibre elongation

between the spinneret/needle and the collector. This phenomenon can be explained by

studies where both strength and elongation at break of PCL composite were found to

decrease significantly with increased filler phase [195,196]. More importantly, the

incorporated inorganic filler phase acts as nucleation agents in the PCL matrix and

noticeably reduce its crystallization half time [197]. These material characterizations

match our experimental observation where the melt-electrospun PCL composite fibres

either broke between the needle tip and collector or solidified before they can attach

to the collector or existing fibres.

Table 5.3 Web of Science search results of ‘melt-electrospinning’ AND ‘polymer composite’ on 10/10/2016

Authors Composite material Year

1. Brown et al. Review paper 2016

2. Lee et al. PCL of different molecular weight 2016

3. Li et al. N/A 2016

4. Kim et al. Silk fibroin/PCL 2015

5. Ren et al. * PCL/SrBG 2014

6. Cao et al. * Multiwalled carbon nanotube/polypropylene 2014

7. Cao et al. Styrene-Acryllonitrile/Siotactic Polypropylene 2013

8. Yoon et al. Nano-/microfibrous poly(L- lactic acid) 2013

9. Carroll et al. Poly(ethylene oxide)/water 2008

10. McCann et al. TiO2-PVP/octadecane 2006

Previously, hybrid solution-electrospinning has been reported in studies where

an elevated temperature was used to assist solution-electrospinning by improving

polymer solubility in the solvents, which were essentially solution-electrospinning

[83,97,98]. In this project, we introduced the concept of hybrid melt-electrospinning

Page 114: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

96 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

to successfully to circumvent the problems associated with using PCL and SrBG

particles and produce continuous fibres of PCL/SrBG composite. Different to the

reported hybrid system which was essentially solution-electrospinning, the hybrid

melt-electrospinning developed in this thesis used a minimal amount of solvent to

assist the electrospinning of polymer composite at melt temperature, which was a melt-

electrospinning process assisted by the solvent. Several key parameters play important

roles in this process: heating temperature, needle tip to collector distance, feeding

speed of the collector, voltage and flow rate of polymer composite. Most of these

parameters interact with each other and changing one individual parameter would

affect the whole scaffold production. The amount of chloroform was optimised to

enable a stable supply of composite material coming out from the electrospinning

needle and the heating temperature was kept at 60 ºC which is below the boiling point

of chloroform to avoid fast drying. Adding more chloroform to the composite would

dissolve electrospun fibres on the collector resulting in a non-porous ‘sheet’ structure

while adding less would not allow a continuous flow of composite fibre jet. The

feeding speed of collecting stage had to match the flow rate of polymer composite so

that straight fibres could be obtained. This speed also correlated with the needle tip to

collector distance and voltage. Longer collection distance would result in unstable

fibres and shorter distance would increase the risk of ‘arcing’. The additional reasons

of using minimum chloroform was to minimize its cytotoxic effect and possible impact

of solvent on the SrBG component during the electrospinning process. In this study, I

optimised the melt electrospinning process to produce smooth composite electrospun

fibres and to stack these fibres into ordered structure via machine control of collector

movement. Overall, this technique exhibited demonstrable advantages of not only

preventing breakage of PCL/SrBG composite jets but also maintaining their stability,

enabling the production of composite scaffolds in a direct writing mode, making it

suitable for 3D printing of scaffolds. These features are crucial to forming PCL/SrBG

composites into 3D electrospun scaffolds with controlled and custom layout and

anatomical precision.

As indicated by the SEM images (Figure 5.2), the PCL/SrBG scaffold fibres

were continuous and followed the machine code (G code) to form a cross-hatch

laydown pattern. However, the PCL/SrBG composite fibres (d=75±21 μm) were larger

in diameter compared to PCL ones (d=28±4 μm). This may have resulted from a

Page 115: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 97

relatively short needle tip to collector distance, not allowing enough space for fibre

elongation. The short collection distance was adopted for depositing straight fibres in

the stability jet region by avoiding ‘whipping’ in the instability region. The addition

of SrBG particles increased the stiffness of PCL fibres mainly due to two reasons: the

composite scaffolds have thicker fibres; and the SrBG filler phase reinforced the fibre

and thus the whole matrix. In addition, the PCL/SrBG fibres were covered by ‘brush’

structures which are approximately 5 μm in length and 0.4 μm in width. These

individual brushes are actually short submicron fibres produced during scaffold

fabrication process due to rapid solvent evaporation and static charge at the surface of

fibres [198]. As there was only minimum amount of chloroform and the viscoelastic

properties of 50% PCL/SrBG composite, the jets solidified before full fibres could be

formed. Such multi-level structures of composite scaffold could potentially provide

increased surface area and additional attachment sites for cells.

The SrBG particles were homogeneously distributed along the scaffold fibres as

observed by confocal microscopy, indicating the composite preparation method to be

effective (Figure 5.3). However, only the surface SrBG particles were shown by this

technique due to the limited penetration of Alizarin red dye into PCL/SrBG composite

fibres. To observe the SrBG particles distribution further deep within the composite

fibres, SEM in the BSE mode was employed to investigate the composite scaffold.

Because the BSE signal intensity is closely related to the atomic number of elements,

BSE images can be used to provide information of elemental distribution in the

specimen [199]. BSE has also been used to identify mineral contents of biological

specimens including bone and teeth [199,200]. The PCL/SrBG scaffolds were

embedded in resin and ground to creat a cross section which exposed SrBG particles

inside the PCL fibres. The cross sections of PCL/SrBG scaffolds were surface scanned

to qualitatively identify SrBG particle distribution as they are brighter than PCL matrix

(Figure 5.4). A Sr mapping was applied on the zoomed-in areas of bright spots to

confirm they were SrBG particles which homogeneously distributed across the whole

scaffold (Figure 5.4). The large areas of SrBG particles indicated high loading of SrBG

filler phase in the composite scaffolds, and it is essential to assess whether the

increased SrBG content could enhance the bioactivity of these scaffolds.

The bioactivity of PCL/SrBG scaffolds relies on the SrBG filler phase, through

1) the formation of CaP layer which plays an important role in cell/tissue attachment

Page 116: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

98 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

to scaffolds on the surface and 2) Si4+ and Sr2+ ion dissolution into the surrounding

environment that synergistically stimulates osteoblast differentiation [58]. Therefore I

found it essential to examine the formation of the surface CaP layer precipitation and

Si4+ and Sr2+ dissolution of the composite scaffolds. In order to determine the

bioactivity, the composite scaffolds were carefully characterised by EDX for surface

CaP deposition and by ICP-MS for ion dissolution α-MEM media. The elemental

concentration PCL/SrBG scaffolds in α-MEM media during a period of 4 weeks

showed a similar pattern to that of bare SrBG particles [58]. The sharp decrease of Ca

and P concentration after 3 h immersion of PCL/SrBG scaffolds (Figure 5.6a and b)

indicated a fast formation of a CaP layer, which was detected on scaffold surface by

EDX analysis (Figure 5.7). Compared to our previous study on 10% PCL/SrBG

scaffolds [191], the formation of CaP layer on 50% PCL/SrBG scaffolds was more

rapid attributed to the increased amount of SrBG within the scaffold. The increase of

Ca and P elements correlated well with the decrease of Ca and P elements in the media

shown in Figure 5.6a and 5.6b. Further studies of CaP precipitation of SrBG should be

carried out in the future as it is reported that the Sr can potentially substitute Ca in the

CaP precipitation process. Additionally, the elemental concentration of metal ions

such as Mg, Sr and Mn could negatively affect the formation of CaP layer [201], which

should also be studied to determine the optimal concentration of Sr2+ in the media.

Furthermore, Sr and Si concentration in the PCL/SrBG dissolution media increased

over the incubation period and the peak values of which were comparable to that of

bare SrBG particles [58]. In the study of SrBG particles with 50% and 100% Sr

substitution [58], the ion dissolution showed a burst release and the concentration of

Sr peaked after 60 min of incubation at around 50 ppm and 85 ppm respectively.

Unfortunately, the ion release profile of SrBG with 75% Sr substitution was not found

in published studies but it is projected to be between the peak values of 100% and 50%

SrBG. According to this analysis, the ion dissolution of Sr showed a burst release of

elements and peaks were reached after 120 min of incubation. In this study, the

concentration of Sr and Si increased sharply after 2 days and 7 days of incubation. This

2-stage increase of ion release may result from the partial degradation of the PCL layer

on the surface due to the high alkaline environment near the surface. The degradation

of PCL then leads to the exposure of more SrBG particles, which released more Si and

Sr ions into the media. When compared to a recent study on PCL/SrBG composite

scaffold [77], the electrospun PCL/SrBG scaffolds in this project released Sr and Si

Page 117: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 99

into the cell culture media with sufficient concentration to potentially stimulate

osteoblast differentiation. Based on the SrBG composition [57], approximately 5.3

wt% of total the amount of Sr element and 5.5 wt% of Si element within the composite

scaffolds were calculated to have released into the media after 28 days. Therefore, the

PCL/SrBG scaffolds had the potential to enable a long-term sustainable ion dissolution

which is desirable for bone regeneration. To confirm the effect of increased SrBG

contents, an in vitro cell study was carried out following the scaffold characterization.

After being fabricated, the PCL/SrBG scaffolds were etched with 5M NaOH for

15 min to enhance surface hydrophilicity and to maximize the exposure of SrBG

particles on the scaffold surface. The etch time in this study was shorter compared to

the 1 hour NaOH etching in study 1 to maintain the structural integrity of the

PCL/SrBG scaffolds. In the PCL/SrBG (50 wt%) scaffolds there were a significant

increase of SrBG filler phase (from 10 wt% to 50 wt%) and removing too much PCL

from the PCL/SrBG fibres would weaken the bonding between fibres at the

intersection of the cross-hatch structure. The PCL scaffolds did not have this concern

but were etched in the same condition as control. As reported previously, an initial pH

increase was observed when the PCL/SrBG scaffolds were immersed in cell culture

media [77], which may pose cytotoxic effect on the seeded MC3T3 cells. The pH

increase was due to the rapid ion exchange of Na+, Sr2+ or Ca2+ in SrBG with H+ from

the cell culture media, and the pH would stop increasing when the Si-OH groups

condensate and repolymerise on the scaffold surface [54]. Therefore, the sterile

PCL/SrBG scaffolds were conditioned in serum-free media for 1 week prior to cell

seeding. The conditioned scaffolds were sterilized again by UV radiation immediately

before cell seeding. Again, it was not necessary for the PCL scaffolds but they were

processed with the same conditions as control.

The LIVE/DEAD assay found only negligible dead cells (red) on both

PCL/SrBG and PCL scaffolds at day 1 and day 7 time points. The live cells (green)

were evenly distributed across the scaffold, which suggested that the increased amount

of SrBG content did not have cytotoxic effects on cells. The stain at the day 7 time

point showed a large number of cells on both types of scaffolds, suggesting that

attachment and migration of MC3T3 cells were not significantly altered by the

incorporation of SrBG to the PCL component, or the increase in fibre diameter (Figure

5.7). The SEM images of cells on PCL/SrBG and PCL scaffolds were in line with the

Page 118: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

100 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

LIVE/DEAD assay results, showing that the scaffold fibres were covered by the

attached cells (Figure 5.8). The active attempt to bridge adjacent scaffold fibres by

MC3T3 cells was observed after 7 days in cell culture at the intersection of the cross-

hatch structures (Figure 5.8b and d). These observations were quantitatively confirmed

by the MTT assays results showing the metabolic activity of cells increased over the

cell culture period among all 4 groups, which suggested that the high loading of SrBG

particles did not have any significant negative effect on the proliferation of the cells

upon the scaffolds. The PCL_O group showed a peak of MTT activity at day 21 time

point due to an increase of cell population on those scaffolds. This could be explained

that the majority of the cells were at the growth/proliferation stage of the osteoblast

development [202]. As the following osteoblasts differentiation took place, the MTT

showed a decrease at the day 28 time point. The interaction between cells and SrBG

particles were again detected via CLSM (Figure 5.9), and cells were found to attach

and well-stretched on the PCL/SrBG scaffold surface with a large amount of SrBG

particles. This further proved that the SrBG filler phase was not cytotoxic to the

attached MC3T3 cells.

Alizarin red S stain, the most commonly used in vitro assays to determine ECM

mineralization [156], was not used in this study as the large amount of SrBG contents

could be stained and interfere with the results. ALP assay was once again used to

quantify the extent of osteoblast differentiation between the experiment groups. As

mentioned earlier, ALP is a known early marker for osteoblast differentiation, the ALP

activity can be used to determine extent of osteoblast differentiation and mineralization

[203]. To exclude the effect of difference in cell numbers on total ALP activity across

all experimental groups, the ALP activity was normalized by the cell numbers

calculated from the PicoGreen DNA quantitation assay. The normalized ALP activity

results indicated that both osteogenic media and the PCL/SrBG scaffolds had

significant impact on the level of ALP activity. More specifically, the cells cultured on

PCL/SrBG scaffolds showed higher ALP activity compared to those on PCL ones

irrespective of culture media types at detection points over the culture period; cells

cultured in osteogenic media showed higher ALP activity compared to those in non-

osteogenic (growth) media (Figure 5.11). Notably, after 21 days in the control growth

media groups, the significantly higher ALP activity of cells on PCL/SrBG scaffolds

compared to cells on PCL scaffolds indicated that the incorporation of 50 wt% SrBG

Page 119: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 101

made the composite scaffolds osteogenic in vitro without osteogenic cytokines. The

normalized ALP activity of PCL_O group at day 21 time point was lower than that of

the day 14 time point because the increase of cell population which matched the cell

metabolic activity assay result. After 28 days, the ALP activity of cells on PCL/SrBG

scaffold in osteogenic media was significantly higher compared to both PCL_O and

PCL/SrBG_C groups, which indicated that in addition to SrBG components, the

osteogenic media was still playing an important role in the osteoblastic differentiation

of MC3T3 cells. When comparing with study 1 (10% SrBG), the increase of SrBG

loading from 10 wt% to 50wt % in the PCL/SrBG composite scaffolds led to drastic

increase of Si4+ and Sr2+ ion concentration: peak concentration of Si4+ almost doubled

from 32.74ppm to 57.57ppm; peak concentration of Sr2+ increased from 55.4ppm to

74.34ppm (Figure 5.5). The increased SrBG led to sufficient Si4+ and Sr2+ ions

available for cells cultured on the PCL/SrBG scaffolds and the synergistic function of

these two ions was key in promoting osteoblastic differentiation as reported previously

[58]. More specifically, the Si4+ showed a more rapid release reaching the peak value

after 14 days of immersion in cell culture media compared to Sr2+ which peaked after

28 days of dissolution (Figure 5.5). This unsynchronized ion release combined with

the fact that the media were changed every two days had contributed to the significant

effect of the PCL/SrBG scaffolds was only observed towards the end of the culture

period.

In order to further support the osteogenic effect of the PCL/SrBG scaffolds, the

expression of ALP gene was investigated (Figure 5.12a). Overall, the ALP gene

expression followed a similar trend of ALP activity where the PCL/SrBG_O showed

the highest ALP expression over the culture period and both scaffold type and culture

media type played important roles in the expression of ALP gene. Notably at the day

21 time point, the ALP gene expression was significantly higher when cells were

cultured on the PCL/SrBG scaffolds group compared to the cells on PCL scaffolds in

both growth control media and media supplemented with osteogenic reagents (ascorbic

acid, β-glycerophosphate and dexamethasone). These discoveries agreed well with

ALP activity at the same time point, which further confirmed the osteogenic capacity

of the PCL/SrBG composite scaffolds, and are in line with the findings of previous

studies by Poh et al.[77] and Gentleman et al.[58]. Osteopontin (OPN), a downstream

gene associated with osteoblast development, was also assessed for its mRNA

Page 120: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

102 Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode

expression in the 4 experimental groups (Figure 5.12b). At day 21 and 28 time points,

upregulation of OPN genes was observed in PCL/SrBG_C and PCL/SrBG_O groups

compared to PCL_C and PCL_O groups respectively. At day 28, the PCL/SrBG_O

group had significantly higher OPN expression compared to PCL_O and PCL_C

groups, which indicated the osteogenic effect of SrBG components of the composite

scaffolds. The OPN expression was also much higher in the PCL/SrBG_C group when

compared to PCL_C group at day 28 (the mean value of PCL/SrBG_C group was over

800 times of the PCL_C group), however, the difference was not statistically

significant. The enhanced ALP activity and upregulation of ALP and OPN genes

indicated that the increased SrBG components did contribute to the osteoinductivity of

the PCL/SrBG composite scaffolds.

5.5 CONCLUSION OF STUDY 2

In order to produce PCL/SrBG composite scaffolds with sufficient SrBG filler

phase, a hybrid melt-electrospinning technique was developed and reported for the

first time in this study. 50% PCL/SrBG composites were successfully fabricated with

this electrospinning technique in a direct writing mode into controlled porosity and

laydown pattern. The SrBG particles were found evenly distributed across the scaffold

fibres. The in vitro bioactivity of the PCL/SrBG scaffolds was characterised and the

concentration of Sr and Si elements in the cell culture media increased over the

incubation period and the peak value of which were comparable to that of bare SrBG

particles. The high concentration of Sr and Si and the sustained release of these

elements indicate that the PCL/SrBG scaffolds can potentially stimulate osteoblast

differentiation. The precipitation of CaP which facilitates cell attachment was

evidenced on the PCL/SrBG scaffold surface, and Ca and P were found to increase

over the incubation time. These results show the hybrid melt-electrospinning technique

is promising for the fabrication of a large variety of polymer/inorganic particle

composite. The PCL/SrBG (50 wt%) scaffolds demonstrated enhanced in vitro

bioactivity compared to PCL/SrBG of 10 wt% SrBG loading (figure 5.13). The

subsequent in vitro cell study on the new scaffolds proved the PCL/SrBG (50 wt%)

scaffolds presented minimum cytotoxicity and showed the ability to support MC3T3

cell attachment and proliferation using LIVE/DEAD assay, MTT assay, SEM and

CLSM imaging techniques. More importantly, the findings in this study also

quantitatively validated the osteogenic capacity of PCL/SrBG scaffolds to stimulate

Page 121: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 5: (Study 2) Developing 50 wt% Strontium-substituted bioactive glass and Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a direct writing mode 103

osteoblast differentiation without osteogenic supplement in the cell culture media.

These properties of PCL/SrBG composite scaffolds thus make them potential tissue

engineered bone substitutes for treating human bone defects.

Figure 5.13 The direct comparison of Si4+ and Sr2+ ion concentration of PCL/SrBG (10wt%) scaffolds in study 1 and PCL/SrBG (50wt%) scaffolds in study 2. The figures indicated increased Si4+ and Sr2+ ion concentration (red lines) of the PCL/SrBG (50wt%) scaffolds dissolution media compared to their concentration in PCL/SrBG (10wt%) scaffolds dissolution media (green lines). The ion concentration of PCL scaffolds dissolution media was examined as controls (blue lines).

Page 122: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 123: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 105

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

A typical scaffold development process includes scaffold fabrication, in vitro

assessment and in vivo assessment with histology being one of the key techniques for

scaffold explant analysis. This PhD project aims to develop a PCL/SrBG composite

scaffold through melt-electrospinning and assess the osteogenic capacity of the

scaffold using in vitro assays and examine their in vivo osteogenesis via animal

implantation followed by explantation analysis using histology. This histology

optimisation study was run in parallel to study 1 and 2 aiming to develop the optimal

histological techniques for the analysis of PCL/SrBG scaffolds at the time of their

implantation. The histological technique optimization was carried out with four animal

species to make sure it would cover the intended animal models for PCL/SrBG

scaffold implantation and also lead to a comprehensive histological study of bone

tissues. Owing to time restrains we did not implant the scaffolds but these will be

implanted in the future and can be assessed using the optimised histology methods

developed in this chapter.

6.1 INTRODUCTION

In recent years, the tissue engineering (TE) research has grown tremendously,

especially with the development of three-dimensional scaffolds for tissue or organ

regeneration [32]. To be able to translate TE scaffolds into clinical applications, a

series of bioactivity tests must be undertaken in vitro and in vivo [100]. Currently, it is

agreed that pre-clinical animal models reflect the complex clinical situation and

provide a real understanding of the tissue regenerative capacity of the

implants/scaffolds [100,101]. However, these in vivo studies are considerably resource

and time intensive. In addition, considering the limited numbers of animals that can be

ethically approved to use, the specimens obtained from these in vivo studies are

extremely precious. Therefore, it is imperative to employ the most appropriate

assessment techniques to obtain maximum information from extremely limited

Page 124: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

106 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

resources. For bone tissue engineers, histology is one of the key ex vivo assessment

techniques to characterise bone formation and mineralization mechanisms including

cellular pattern and distribution within the bone matrix [105].

Histology is traditionally an analytical method in biology and medicine for the

structure and composition of tissues at the microscopic level [6]. It is also widely

employed in bone TE research as a primary method of investigation, as well as a

validation of other analytical techniques, such as conventional x-rays [204] and micro-

computed tomography imaging [205]. By processing tissue explants into thin sections

on the micron scale through fixation, embedding, sectioning and staining steps,

histology provides microscopic analysis of “two-dimensional” tissue sections [109].

Histology techniques are highly versatile with numerous stains available for specific

tissue types. Despite the recent advances in histological techniques, it remains a

challenge to process mineralized tissues such as bone and teeth due to their

significantly greater hardness and the heterogeneity of the tissues in healing bones, and

requires the use of different embedding materials than paraffin which remains the most

widely used embedding material for soft tissues [8].

In order to section bulk bone explants, two approaches have been employed

which aim to match the hardness of bone to that of the embedding media: i) to soften

bone specimens by decalcification (by reagents such as ethylenediaminetetraacetic

acid (EDTA)) for paraffin embedding; and ii) to use hard embedding materials such as

plastic/resin for non-decalcified bones [109]. The embedding media and sectioning

techniques for producing bone tissue sections are summarized in table 6.1. Paraffin

embedded specimens are routinely sectioned by a rotary microtome to yield good bone

morphology, enzyme, and immunohistochemistry results through standard histological

stains [6,109]. This approach, however, can lead to poor preservation of bone structure

[110] and often loses information on bone regeneration and mineralization due to the

decalcification process. Decalcification of bone is also a time-consuming process that

can take several months. Other prominent disadvantages of paraffin embedding

approaches include limited specimen size and inability to process metallic implants.

Page 125: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 107

Table 6.1 Summary of embedding media and sectioning techniques in study 3. Embedding media Paraffin MMA resin Technovit 9100 resin

Bone tissue state Decalcified Non-decalcified Non-decalcified

Sectioning

technique

Paraffin

microtome

Sledge

microtome

Ground

sectioning

Sledge

microtome

Ground

sectioning

Section naming

and thickness

Paraffin

sections ~ 5

μm

Resin thin

sections ~

10 μm

Resin ground

sections ~ 20 to

50 μm

Resin thin

sections ~

10 μm

Resin ground

sections ~ 20 to

50 μm

As bone tissue engineering research advances towards larger and more complex

tissues, the traditional paraffin embedding approach can no longer provide optimal

results, especially at the scaffold/tissue or soft/hard tissue interfaces. Plastic

embedding, most commonly methyl methacrylate (MMA) resin, overcomes the

shortfalls with paraffin approach and causes minimum disruption of tissue and cellular

morphology. The MMA embedding approach has been providing quality

morphological results in hard tissue research for nearly 50 years [8,110,122].

However, MMA sections are not appropriate for protein markers detection techniques

like IHC owing to destruction of enzymes and antigen epitopes during the exothermic

polymerization process (reaction temperature up to 80 °C) [121]. To overcome this

disadvantage of conventional MMA, low temperature polymerizing resin was later

introduced [8,110]. The Technovit 9100 New® resin polymerizes at a temperature as

low as -20 °C [8,110,121,123], which allows the preservation of epitopes that are

suitable for IHC examination. Both MMA and Technovit 9100 New® resin embedded

tissues can be sectioned with ground sectioning technique and heavy-duty sledge

microtomes, in the same way as traditional MMA [109].

Haematoxylin and eosin (H&E) stain and IHC are the most routine histological

stains that demonstrate general tissue morphology and osteogenic/angiogenic markers

respectively. Other more specific stains differentiate mineralized bone from

surrounding tissues, including von Kossa silver nitrate methods and Goldner’s

trichrome stains [109]. Goldner’s trichrome stain being one of the most popular

staining technique that has been employed in numerous in vivo bone regeneration

studies [8-26].

Page 126: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

108 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

With all of these histological techniques available to TE researchers, the

selection of techniques is, however, largely based on individual experience and can

lead to suboptimal results or irreversible loss of information if explanted specimens

are not appropriately and reproducibly processed. Over the years, different research

groups have been publishing results of histological stains of bone specimens using

their own variation of standard operating protocols. Without agreed standards with

respect to what constitutes successful histological staining, it is difficult to

communicate and compare the progress of bone mineralization based on the different

staining results from various research groups. As mentioned above, Goldner’s

trichrome has been one of most important histological stains for bone specimens, and

images of bone/bone implant sections stained by this method from the literatures have

demonstrated inconsistent and uneven stains of mineralized bone and surrounding

connective tissues [125,131,132,206]. Some of the exemplary images can be found in

Appendix A.

Presently, no published data exists to directly compare the staining results of

bone specimens embedded in paraffin, MMA, and Technovit 9100 NEW®. In this

study, we are therefore filling in this gap by optimizing and comparing the staining

techniques for the assessment of mineralized tissue processed via above mentioned

embedding and sectioning approaches using various common animal tissues including

sheep tibia, pig fibula, rat and mouse legs and mouse paws. The full panels of

microscopic images of stains are provided with detailed operating procedures, which

are expected to standardize the histological techniques for bone specimen analysis.

Here I also demonstrate in fine details of the methodology to divide and distribute

large tissue-engineering bone specimens for both resin and paraffin histology for their

comprehensive analysis. The outcome of this study can be used as a guide to

researchers in bone TE field for their pre-planning of histological analysis based on

specific research aims by providing a series of optimised staining methods which are

optimised for different animal species.

6.2 MATERIALS AND METHODS

6.2.1 Bone tissues and pre-processing preparation

In this study, I utilised bone tissues across four animal species: sheep tibiae

[207], pig fibulae [208], mouse paws and legs [209], rat legs [102], as well as, tissue

Page 127: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 109

engineered sheep tibiae with polymeric and titanium implants. The use of bone tissues

and bone explants were approved by Queensland University of Technology Animal

Ethics Committee (ethics approval numbers: 1400000776 and 1000001139).

The study design is summarized in Figure 6.1. Prior to fixation, samples were

resected to remove excess soft tissue. Triplicate 10 mm thick transverse sections were

cut for sheep tibia, 5 mm transverse sections for pig fibulae, 3 mm thick for mouse and

rat legs were cut using an EXAKT 310 Diamond Band Saw (EXAKT Apparatebau

GmbH & Co.KG, Norderstedt, Germany). Four mouse paws were kept as an entire

sample measuring between 1.4 - 1.9 cm. The tissue engineered bone explants were cut

as illustrated in Figure 6.4. The specimens were fixed in 10% neutral buffered formalin

(10:1 volume of fixative to tissue) for 24 h in sealed containers. To completely remove

the fixative solution, the bone specimens were thoroughly washed in deionized water

(DI water) for 4 h prior to further processing. Bone specimen triplicates were divided

equally into three groups for embedding in three media according to histological

methodology: paraffin, Methyl Methacrylate (MMA) and Technovit 9100 New®.

Figure 6.1 Schematic summary of study 3 design. The animal tissues were divided into three identical parts and embedded in paraffin, MMA and Technovit 9100 resin respectively. The obtained sections of these tissues were then stained by H&E, Von Kossa, Goldner’s trichrome and IHC techniques. The staining results were then compared.

6.2.2 Processing and embedding

Paraffin. Bone tissues were decalcified in 10% EDTA (Ajax Finechem,

Australia) at 4 °C for 14 weeks to ensure complete demineralization and the endpoint

Page 128: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

110 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

was carefully determined by probing the tissues with a sharp needle. EDTA solution

was refreshed weekly. The bone specimens were briefly rinsed with DI water to

remove excess of EDTA solution, and then placed in embedding cassettes (Techno

Plas, South Australia) for processing in an ExcelsiorTM ES Tissue Processor (Thermo

Scientific, Australia), prior to embedding in molten paraffin wax at 60 °C (Thermo

Shandon Histocentre 3 Embedding Station, Thermo Scientific, Australia).

Resin. Specimens processed for MMA and Technovit resin were dehydrated for

3 weeks through graded series of ascending concentrations of ethanol. Specimens were

then degreased in 2 changes of xylene for 8 h at RT to remove ethanol residual and

facilitate resin penetration.

Methyl Methacrylate (MMA) Following degreasing with xylene, the specimens

were immersed in MMA infiltration solution (MMA supplemented with 3%

polyethylene glycol (PEG)) of 20 times of volume to that of tissues and kept for 1-2

weeks at RT: sheep tibiae, pig fibulae and mouse paws for 2 weeks and rat and mouse

legs for 1 week. The base molds were prepared during the infiltration by polymerizing

approximately 1 cm of MMA embedding solution (MMA+3% PEG+0.3% di(4-tert-

butylcyclohexyl) peroxydicarbonate Perkadox, Akzo Nobel Polymer Chemicals LLC)

at the bottom of embedding containers. Specimens were then transferred into the base

molds and covered by MMA embedding solution to 1-2 cm above the top of

specimens. The embedded specimens were vacuumed at -70 KPa for 2-5 min to

remove oxygen and polymerized at RT for 3-5 days.

Technovit 9100 New® Following degreasing with xylene, the allocated

specimens for Technovit 9100 New® methodology were processed and embedded in

the low-temperature embedding system Technovit 9100 New® (Heraeus Kulzer

GmbH, Germany). Pre-infiltration, infiltration, and embedding solutions were

prepared according to Table 6.1, using Technovit 9100 NEW® basic solution

destabilized by filtering through chromatography column loaded with 50 g of

aluminium oxide. All bone specimens were immersed in pre-infiltration solution at 4

°C for 1 week. Following pre-infiltration, the specimens were transferred into the

infiltration solution and vacuumed at -70 KPa for 2-5 min to facilitate infiltration. The

specimens were stored in infiltration solution at 4 °C for 1 week. The pre-infiltration

and infiltration solutions were 20 times of volume to that of tissues being processed.

The infiltrated specimens were finally transferred into plastic embedding moulds,

Page 129: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 111

filled with fresh embedding solution (a mixture of 9 parts of stock solution A and 1

part of stock solution B) and vacuumed at -70 KPa for 2-5 min to remove oxygen. The

polymerization took place at -20 °C for 5-7 days. Once polymerized, the resin blocks

were transferred to 4 °C for 2 h, before storage at RT.

Table 6.2 Preparation of Technovit 9100 New® solutions

Solutions Basic solution

PMMA powder Hardener 1 Hardener 2 Regulator Storage

Pre-infiltration 200 ml 1 g 4 °C Infiltration 250 ml 20 g 1 g 4 °C Stock solution A* 500 ml 80 g 3 g 4 °C Stock solution B* 50 ml 4 ml 2 ml 4 °C

* Solution A and Solution B are polymerization solutions which must be mixed up in the ratio of 9A:1B immediately prior to use.

6.2.3 Sectioning

Paraffin microtomy. Five-micron transverse tissue sections were generated on

a Leica RM2235 rotary microtome (Leica Biosystems, Nussloch Germany). The

ribbons of sections were floated on a 40 °C water bath to unfold and collected onto

polylysine-coated microscope slides (Thermo Scientific, Australia). To improve

sectioning quality, the paraffin blocks were cooled on ice for 5 min after every 3-4

ribbons. The slides were oven dried at 60 °C for 16 h.

Resin sledge microtomy. Tissue engineered sheep tibiae embedded in either

MMA or Technovit 9100 were sectioned with a sledge microtome (Polycut-S,

Reichert-Jung, International Medical Equipment, USA) using a tungsten carbide blade

at a section thickness of 8 μm. The resin blocks were moistened constantly with ethanol

(70% for MMA and 30% for Technovit) to improve section quality. Sections were then

flattened with 95% ethanol onto gelatin-coated microscope slides, covered with

polyethylene film and compressed between bibulous paper sheets to remove excess

ethanol. Finally, the slides were stacked in a metal slide holder under compression and

dried in an oven at 60 °C for 3-4 days. The resin sections obtained by the sledge

microtome are referred to as resin thin sections in this paper to distinguish them from

resin ground sections, which are much thicker.

Resin ground sectioning. As described previously [111,210], this technique

involves two major steps: cutting a section from the polymerized resin block and

grinding the section to an appropriate thickness. Methyl methacrylate and Technovit

Page 130: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

112 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

polymerized resin blocks were shaped and trimmed of excess resin, leaving a border

of resin 5 mm from the specimen, using an Exakt 310 diamond band saw (EXAKT

Advanced Technologies GmbH, Germany). An overview of the process for resin block

preparation before sectioning is schematically shown in Figure 6.2. Blocks were glued

to a 50 x 100 x 2mm acrylic slide (A) with cold curing resin Technovit 4000 system

(Technovit powder mixed in Technovit syrup I and syrup II, EXAKT Advanced

Technologies GmbH, Germany) using the Exakt vacuum system 401 (EXAKT

Advanced Technologies GmbH, Germany). The to-be-sectioned surface of resin block

was polished with 800 grit sandpaper to a smooth surface, parallel to slide A with the

Exakt 400 CS microgrinding system (EXAKT Advanced Technologies GmbH,

Germany). The resin block surface was then degreased with ethanol (100% ethanol for

MMA and 30% ethanol for Technovit embedded blocks) and affixed onto a second

acrylic slide (B) (dimension: 50 × 100 × 2 mm) with Technovit 7210 VLC photo curing

adhesive (EXAKT Advanced Technologies GmbH, Germany).

Page 131: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 113

Figure 6.2 An overview of resin block preparation and ground sectioning process. The tissues embedded in resin are processed following the direction of arrows for the preparation of double-slide ‘sandwich’ structure. The double-slide prepared resin blocks are cut by the saw along the black line and processed following the direction of arrows for the production of resin ground sections. The resin ground sections are then stained by various histological staining techniques as detailed in section 6.2.4.

The overview of cutting and grinding of the prepared sandwiches structure is

shown in Figure 6.2. The mounted resin block was then cut with the Exakt 310

diamond band saw producing a section 100 μm – 200 μm thick. To yield the desired

thickness, the amount of tissue and resin to be removed was pre-selected with the AW

110 electronic measuring and control System (EXAKT Advanced Technologies

GmbH, Germany) and posteriorly ground using an Exakt 400 CS microgrinding

system (EXAKT Advanced Technologies GmbH, Germany) guided by a sequence of

finer abrasive papers summarized in Table 6.3. For sheep tibiae, pig fibulae, rat legs,

Page 132: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

114 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

mouse paws, and legs bones sections were ground down to ~27 μm while tissue-

engineered sheep tibiae sections were ground down to a final thickness of ~50 μm. The

thickness measurement was carried out with a micrometer (Mitutoyo, USA). After the

desired section thickness was reached, specimens were air dried at RT prior to staining.

Table 6.3 Suitable sandpaper grits according to section thickness: a. final thickness=27 μm; b. final thickness=50 μm

a Thickness of Sample Grit of Paper Amount to be removed

250 microns P800 130 microns 120 microns P1000 40 microns 80 microns P1200 40 microns 40 microns P2500 10 microns 30 microns P4000 3 microns

Final section 27 microns

b

Thickness of Sample Grit of Paper Amount to be removed 250 microns P800 97 microns 153 microns P1000 40 microns 103 microns P1200 40 microns 63 microns P2500 10 microns 53 microns P4000 3 microns

Final section 50 microns

6.2.4 Staining

The majority of conventional histological stains have been developed for

paraffin sections. Here, I describe optimised and standardized staining protocols for

resin thin and ground sections.

Haematoxylin & Eosin (H&E) staining

H&E is one the principle stains in histology that allows a general overview of

the histological sections. The nuclei of cells are stained blue by hematoxylin and the

eosin stains the cytoplasm pink [109].

Paraffin. The sections were deparaffinized with 2 changes of xylene (8 min

each) and rehydrated with descending concentrations of ethanol (100%, 90% and 70%,

2 min each), and placed in DI water for 5 min prior to staining. The sections were

Page 133: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 115

stained with Mayer’s haematoxylin (CliniPure, Grale HDS, Australia) for 10 min at

RT, followed by 10 min blueing under warm running tap water. The sections were then

stained with eosin (CliniPure, Grale HDS, Australia) for 10 s at RT. Following

staining, the sections were dehydrated in ascending concentrations of ethanol and

mounted with Eukitt mounting media (Sigma, Australia).

Resin. The staining protocol for resin sections was optimised during this PhD

project. Both MMA and Technovit ground sections were surface etched by immersion

in xylene for 20 min followed by immersion in 100% ethanol for 20 min. Following

rehydration with a descending concentration of ethanol (90% and 70%, 5 min each),

the sections were placed in DI water for 5 min prior to staining. The resin ground

sections were stained with Weigert’s haematoxylin for 15 min, followed by 10 min

blueing under warm running tap water, differentiated in 1% acetic acid (2 × 1 min)

(Merck, Australia), and then stained with eosin for 30 s. The sections were then air-

dried, cleared in xylene and mounted with Eukitt mounting media (Sigma, Australia).

Goldner’s trichrome staining

Goldner’s trichrome staining is a commonly used method in bone histology,

allowing tissue differentiation based on specific colours binding to different tissue and

cell structures providing colour difference for each tissue structure as well as by

morphological identification [109].

Goldner’s trichrome staining procedures were optimised based on existing

standard operating procedures in our lab. The solutions for Goldner’s trichrome

staining are listed in Table 6.4.

Table 6.4 Goldner’s trichrome staining solutions Solutions Recipe

Weigert’s iron hematoxylin A 10g haematoxylin monohydrate (Merck, Australia)

dissolved in 1000 mL 95% ethanol

Weigert’s iron hematoxylin B 5.8 g ferric chloride (Merck, Australia) and 5 mL

hydrochloric acids dissolved in 500 mL DI water

Acid Fuchsin Ponceau A 6g Ponceau Xylidine (Merck, Australia) dissolved in

600 mL DI water

Acid Fuchsin Ponceau B 2g Acid Fuchsin (Merck, Australia) dissolved in 600

mL DI water

Page 134: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

116 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Tungstophosphoric acid–orange G 10g orange G (Merck, Australia) and 20g

tungstophosphoric acid (Merck, Australia) dissolved

in 500 mL DI water

Light green (SF yellowish) 1g light green SF yellowish (Merck, Australia) and

1mL acetic acid (Merck, Australia) dissolved in 500

mL DI water

Paraffin. 5 μm sections were deparaffinised with 2 changes of xylene (8 min

each) and rehydrated with a descending concentration of ethanol (100%, 90% and

70%, 2 min each), and placed in DI water 5 min prior to staining. The sections were

placed in Weigert’s iron haematoxylin solution for 25 min and blued under warm water

for 10 min. The sections were then immersed in acid Fuchsin-Ponceau working

solution for 10 min, rinsed in fresh 1% acetic acid (2 × 30 s) before being placed in

tungstophosphoric acid - orange G solution for 20 min. Following another 2 × 30 s

rinse in 1% acetic acid, the sections were immersed in the light green solution for 10

minutes. Subsequently, the sections were rinsed in 1% acetic acid (2 × 30 s) and then

rinsed in DI water for 2 min. The stained sections were dehydrated in ascending

concentrations of ethanol (70% and 100%, 2 × 30 s), cleared in xylene (2 × 6 min),

and finally mounted with Eukitt mounting media.

Resin. Resin thin sections (8 μm) were deplasticized in acetone (2 × 20 min),

rehydrated with descending concentrations of ethanol (100%, 90% and 70%, 5 min

each), and placed in DI water for 5 min. The resin ground sections were surface etched

by immersion in xylene and 100% ethanol for 20 min each to facilitate dye infiltration.

Following rehydration with a descending ethanol series (90% and 70 %, 5 min each),

the resin ground sections were placed in DI water for 5 min prior to staining. Both

resin thin and ground sections were placed in Weigert’s iron haematoxylin solution

for 25 min and blued under warm tap water for 10 min. These sections were immersed

in acid Fuchsin-Ponceau working solution for 10 min, rinsed in fresh 1% acetic acid

(2 × 30 s) before being placed in tungstophosphoric acid - orange G solution for 20

min. Following another 2 × 30 s rinse in 1% acetic acid, the sections were immersed

in the light green solution for 10 min (resin thin sections) and 15 min (resin ground

sections). Subsequently, the sections were rinsed in 1% acetic acid (2 × 30 s) and then

rinsed in DI water for 2 min. (Some resin ground sections need extended immersion

Page 135: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 117

in light green solution to stain the mineralized bone sufficiently.) The stained resin

thin sections were dehydrated in ascending concentrations of ethanol (70% and 100%,

2 × 30 s), while the resin ground sections were air dried in the dark for ~1 h. Both

sections were cleared in xylene (2 × 6 min for resin thin sections and 2 x 30 s for resin

ground sections) and finally mounted with Eukitt mounting media.

Von Kossa/MacNeal’s tetrachrome staining

The Von kossa stain is intended for histological visualization of calcium

deposition in sections. When the tissue sections are treated with a silver nitrate solution,

the silver ions are deposited by replacing calcium, following a developing stage, the

silver ions are reduced to metallic silver, which are visualised as a black colour [109].

Von Kossa staining was optimised based on previously described protocols

using solutions shown in Table 6.5 [211–213].

Table 6.5 Von Kossa/MacNeal’ tetrachrome staining solutions Solutions Recipe

5% Silver nitrate 20g Silver Nitrate dissolved in 400 ml DI water, filtered before use

5% sodium carbonate-

formaldehyde

22.5g sodium carbonate dissolved in the mixture of 337.5 mL of DI water

and 112.5 mL of 37% formaldehyde

Farmer’s Diminisher 2g potassium ferricyanide and 40g sodium dissolved in thiosulfate in 420

mL of DI water

Paraffin. Sections were deparaffinised and rehydrated, as previously described.

The sections were immersed in 5% silver nitrate solution for 5 min in the dark.

Following incubation, sections were rinsed in DI water (3 × 1 min) and immersed in a

5% sodium carbonate-formaldehyde solution for 2 min. Following, DI water rinse (2

× 1 min), sections were then submerged in fresh Farmer’s Diminisher solution for 30

s. Sections were then washed under running tap water for 20 min and counterstained

for 10 min with MacNeal’s tetrachrome solution (Dorn and Hart Microedge Inc., US).

After counterstaining, the sections were rinsed with DI water (4 × 30 s), then

dehydrated with ascending concentrations of ethanol (70%, 90% and 100%, 30 s each)

and mounted with Eukitt mounting media.

Page 136: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

118 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Resin. Thin sections were deplasticized in acetone (2 × 20 min), and rehydrated

with a descending ethanol series (100%, 90% and 70%, 5 min each) and placed in DI

water 5 min prior to staining. Resin ground sections were brought into DI water for 5

min prior to staining. Following incubation, sections (both thin and ground) were

rinsed in DI water (3 × 1 min) and immersed in a 5% sodium carbonate-formaldehyde

solution for 2 min. Followed by DI water rinse (2 × 1 min), sections were submerged

in fresh Farmer’s Diminisher solution for 30 s. Sections were then washed under

running tap water for 20 min and counterstained for 10 min with MacNeal’s

tetrachrome solution. After counterstaining, the resin thin sections were rinsed with

DI water (4 × 30 s), then dehydrated with ascending grades of ethanol (70%, 90% and

100%, 30 s each) and mounted with Eukitt mounting media. The resin ground sections

were air dried in the dark, cleared in xylene and mounted.

Immunohistochemistry (IHC)

IHC is used in histology to detect proteins of interest in cells of a tissue section

by exploiting the principle of specific binding between antibodies and antigens [109].

In bone tissue engineering (TE) research, IHC is used to locate specific osteogenic

and/or angiogenic markers on the regenerated bone, which provides information on

for example tissue regeneration progress and formation of new blood vessels. The

information provided by IHC is essential for the assessment of TE strategies.

Paraffin. The sections were deparaffinised with 2 changes of xylene (8 min

each) and rehydrated with descending concentrations of ethanol (100%, 90%, 70%

and 50%, 2 min each), and placed in DI water 5 min prior to staining.

Resin. Ground resin sections were deplasticized in three changes of 2-Methoxyl

Ethylacetate (2-MEA) for 48h as suggested in the literature [214] and rehydrated into

DI water.

Tissue sections were processed following standard IHC procedures, as

previously described [1, 5, 10-13]. In brief, both resin and paraffin sections were

circled by delimiting pen (DAKO, Australia) and slides placed in 50 mM Tris–HCl

buffer (pH 7.4). The endogenous peroxidase activity was blocked by 3% H2O2 in Tris–

HCl for 20 min. After washes (3 x 2 min) with Tris buffer (pH 7.4), sections were

incubated with proteinase K (DAKO, Botany, Australia) for 20 min for antigen

retrieval. The sections were incubated with 2% bovine serum albumin (BSA) (Sigma,

Page 137: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 119

Sydney, Australia) in a humidified chamber at RT for 40 min to block non-specific

binding sites. The sections were incubated overnight at 4 °C with the following

osteogenic markers (diluted in 2% BSA): type I collagen (ColI, 2 μg/ml polyclonal

rabbit, #ab34710, Abcam, UK), osteocalcin (OC, 2 μg/ml polyclonal rabbit, #ab93876,

Abcam, UK) and an endothelial marker von Willebrand Factor (vWF, ready-to-use,

polyclonal rabbit anti human, DAKO Australia). To exclude false positive staining as

a result of rabbit IgG binding non-specifically to Fc receptors, two types of negative

controls were used: rabbit primary antibody isotype control (0.5 μg/ml, #086199,

Invitrogen, Australia) and 2% BSA negative control. Sections were incubated with the

specific antibodies or control solutions in a humidified chamber at 4 °C overnight.

Following 3 × 2 min washes in Tris-HCl buffer (pH 7.4), sections were incubated with

peroxidase-labelled dextran polymer conjugated to goat anti-mouse and anti-rabbit

immunoglobulins (EnVision+ Dual Link System Peroxidase, DAKO) at RT in

humidified chambers for 60 min. The sections were incubated for 10 s with a liquid

3,3-diaminobenzidine (DAB)-based system (DAKO, Australia) for colour

development. All sections were dehydrated in ascending grades of ethanol and

mounted with Eukitt mounting media.

6.2.5 Microscopy and image documentation

Images of histologically stained specimen were acquired and analysed using a

Zeiss Axio Imager M2 light microscope equipped with a scanning stage, Zeiss

AxioCam Mrc digital camera and Zeiss ZEN blue software (Zeiss, Oberkochen,

Germany).

6.3 RESULTS AND DISCUSSION

6.3.1 Study Overview

To ensure the PCL/SrBG composite scaffolds developed in this PhD project

could be effectively evaluated with the endpoint histological analysis, study 3

comprehensively optimised and standardized histological techniques using four

animal species. Following the optimization, I applied the histological processes to

tissue engineered bone specimens. Three most frequently used animal models in bone

tissue engineering research were studied: rat (38%), mouse (13%) and sheep (11%),

accounting for 62% of the total usage [105]. Pig bones were also included in this study

because of its close similarity in composition and biology with human [101]. For

Page 138: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

120 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

histological standardization and comparison, the native bone specimens were

embedded in the representative embedding materials (paraffin, MMA, and Technovit

9100 NEW®) commonly employed in bone histological studies. Other types of

embedding media such as Technovit 7100 (Glycol Methacrylate resin) were not

explored here as they do not provide additional features. Microtome sectioning of non-

demineralized mature cortical bones was found challenging with the existing system

in our lab, therefore, the Exakt ground sectioning system was employed as the main

technique for resin section production. Sledge microtomes were used to section tissue

engineered bone and these sections are referred as resin thin sections (relative to resin

ground sections which are much thicker) in this paper. Four important stains for bone

histology were selected for comparison in this project: H&E for general tissue

morphology; von Kossa for mineralized bone and Goldner’s trichrome for hard/soft

tissue discrimination; and IHC for specific protein markers.

6.3.2 Stain optimization and comparison

Goldner’s trichrome stain is widely used in bone histology because of its ability

to sharply discriminate mature bone matrix to immature new bone matrix and

surrounding soft tissues without losing cellular information [215]. Because of its

excellent cell staining, Goldner’s trichrome is also considered more valuable than other

stains in pathology [109]. However to our knowledge, the currently published staining

protocols provided either sub-optimal or non-repeatable staining results for the resin

ground sections of non-decalcified bone specimens. Therefore here I present our

optimised staining procedure for application of Goldner’s trichrome stain to resin

ground sections of the 4 different species of bone. This stain was proven to be

reproducible across all four animal species providing consistent results, the

representative images of which are presented in Figure 6.3. Overall, good tissue

discrimination was found on resin ground sections where the mature bone matrix

stains green, immature bone matrix stains red, while the surrounding soft tissue stain

orange (Figure 6.3a, b, d, e, g, h, j, k). The cells are visible both in the bone matrix

(osteocytes) and surrounding soft tissues where the nuclei stain dark blue. Osteoid was

observed as a dark red line along the edges of mineralized bone matrix. The osteoblasts

can be found along the osteoid. Additionally, chondrocytes staining is shown as bright

yellow-orange with dark blue nuclei which are clearly visible on the articular joints of

mouse paws (Figure 6.3g, h and i). No significant difference of staining was found

Page 139: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 121

between specimens embedded in MMA and Technovit 9100 NEW®. In comparison,

such details were lost on decalcified paraffin sections (Figure 6.3c, f, i, l), where the

bone, connective tissue and cartilage all stain green with nuclei staining dark blue since

bone mineral contents were completely removed prior to embedding. Across species,

little variance was observed in the staining specificity. The tissue discrimination and

cellular information envisaged by the Goldner’s trichrome stain are essential to

researchers who want to understand bone regeneration and mineralization progress and

the tissue/scaffold interface.

Figure 6.3 Comprehensive comparison of Goldner’s trichrome staining on resin ground sections and paraffin sections of bone of all four animal species. For the ease of comparison, the images are categorized by both animal species: (a,b,c) sheep tibiae, (d,e,f) pig fibulae, (g,h,i) mouse paws, and (j,k,l) rat leg bone; and types of embedding media: (a,d,g,j) MMA, (b,e,h,k) Technovit 9100 New®, and (c,f,i,l) paraffin. The resin ground sections are ~27 µm

Page 140: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

122 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

in thickness, and paraffin sections are 5 µm in thickness. Inserts show overall images of the entire stained tissue. In (a,d,g,j) MMA and (b,e,h,k) Technovit 9100 NEW® ground sections, mineralized bone stains green, connective tissue and cartilage stain orange, osteoid stains red and cell nuclei stain dark blue. While in paraffin sections (c,f,i,l), bone, connective tissue and cartilage stain green, with nuclei staining dark blue. Scale bar = 25 µm

Von Kossa/MacNeal’s tetrachrome stain plays an important role in bone

research and the staining results indicated the discrimination of mineralized bone

(brown/black) and connective tissue (blue) with nuclei (dark blue) on resin ground

sections (Supplementary Figure 6.2). The location of mineralized tissue shown by von

Kossa stain agreed with that shown by Goldner’s trichrome stain in the identical tissue

cut. The colour of mineralized bone was not as densely black as that of resin thin

sections (Figure 6.5b3). This owes to the silver ions only being able to access the

surface layer of the mineralized bone matrix during impregnation as the bottom of

sections were glued to the slide, the existence of resin around the tissue sections further

restricted silver ion penetration and deposition on bone matrix. The cellular details on

the mineralized bone matrix were hardly visible for both thin and ground resin sections

as the whole matrix were covered by black metallic silver. There was no significant

difference in staining between specimens embedded in MMA and Technovit 9100

NEW®. In comparison, the decalcified paraffin sections did not take up any silver ion

and remained pale as there were no calcium ions to be replaced by silver ions on the

decalcified tissues.

In addition to the bone-specific stains, I also examined and compared the most

frequently employed histological stain: the haematoxylin & eosin (H&E) stain on resin

thin and ground sections. I used an existing H&E staining protocol [111] for resin

ground sections and found significant background staining of Mayer’s haematoxylin

on mineralized bone matrix. I hypothesized that it was due to the hemalum of Mayer’s

hematoxylin also binding to the anion groups in the mineral content. I then introduced

differentiating steps using 1% acetic acid to minimize hematoxylin stain on the

mineralized matrix. However, the differentiation decolorized the nuclei stain thus

affected the contrast between cell nuclei and cytoplasm. Mayer’s hematoxylin is an

aluminium modified hematoxylin solution and when exposed to acid it’s rapidly

removed from tissue sections because the aluminium hematoxylin dye-lake formation

fails to happen due to lack of OH− ions [216]. Therefore, I used Weigert’s iron

Page 141: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 123

haematoxylin instead of Mayer’s haematoxylin for its resistance to acidic

differentiating solutions. Different to Mayer’s hematoxylin, Weigert’s iron

hematoxylin uses acidified ferric chloride as the mordant (chemicals combined with

hematoxylin) which can sustain acids [216]. The staining results showed improvement

of contrast between cell nuclei and cytoplasm on resin ground sections but were still

suboptimal compared to H&E staining of paraffin sections (Supplementary Figure

6.1).

Another common histological stain is immunohistochemistry (IHC). Here I used

a published method to deplasticize the resin ground sections [214] and stained both

resin ground sections and paraffin sections using the protocol established in our lab. I

examined two osteogenic markers: collagen type I (ColI) and osteocalcin (OC) and an

angiogenic marker: von Willebrand factor (vWF). Both Technovit 9100 NEW® and

paraffin sections showed immunoreactivity and stained positive for all three antibodies

(Supplementary Figure 6.3). Because of the exothermal nature of MMA

polymerization, the surface epitopes of antigens were destroyed by the high

temperature (80 °C), and as such, specimens embedded in MMA were not

immunoreactive for any of these antibodies and the results were not included in this

thesis. To overcome these difficulties of resin embedded specimens, Technovit 9100

NEW® was designed to preserve the immunoreactivity of specimens by polymerizing

at under-freezing temperature thus prevented the damage of antigens, making it

possible to examine specific markers of undecalcified bone tissues with IHC [110].

Similar staining intensity was found between resin and paraffin sections for ColI

(Supplementary Figure 6.3a1-a8) and OC (Supplementary Figure 6.3b1-b8) across all

4 animal species. More intense staining was found in Technovit 9100 NEW® sections

incubated with vWF antibodies compared to paraffin sections possibly due to greater

tissue thickness of resin ground sections, which provided a better understanding of

blood vessels morphology and their localization within the bone matrix

(Supplementary Figure 6.3ca1-c8). For bone tissue engineers, the immunolabelling of

Technovit 9100 NEW® sections can be valuable in providing the progress of bone

regeneration and mineralization, and more importantly the development and ingrowth

of blood vessels within the bone defect.

Even though paraffin embedded bone specimens do not preserve mineral

contents of bone, researchers have found ways to obtain genetic information from

Page 142: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

124 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

sections of paraffin-embedded tissues. This process involves cell/tissue isolation and

RNA extraction from the isolated tissues. Laser microdissectioning (LMD) technique,

also known as Laser Capture Microdissection (LCM), is a contact- and contamination-

free method for isolating specific single cells or entire areas of tissue from a wide

variety of tissue samples [217]. LMD allows researchers to pick specific area of

interest or specific tissue/cell types within the paraffin section. Several methods have

been developed to extract RNA from the isolated sections, which allows subsequent

analysis of selected tissue/cells using further molecular biological methods such as

PCR and real-time PCR [218]. The quantitative data obtained from the combination of

LMD and subsequent techniques provide important complementary information to the

qualitative histological results. The fact that these techniques allow retrospective

biomarker studies of paraffin sections have made them popular in a large number of

research fields [219–221].

Ground sectioning vs resin microtome sectioning

As shown in Figure 6.4, a slab of tissue engineered sheep tibia was embedded in

Technovit 9100 NEW® and sectioned with both sledge microtome and Exakt cutting

and grinding system (Figure 6.4). The resin thin section showed noticeable 17%

compression in size compared to the original specimen, while resin ground sections

showed no such effect (Figure 6.4c and d). By comparing the sections stained with the

optimised Goldner’s trichrome technique, it is obvious that the resin ground section

not only better preserved the tissue morphology than the resin thin section, but also it

showed better differentiation of mineralized bone and soft tissues (Figure 6.4e). High

magnification images of stained resin ground section showed intact osteon structure

and Haversian canal (Figure 6.4g). In comparison, the resin thin section displayed

poorer overall morphology with shredded mineralized matrix (Figure 6.4f). High

magnification images showed compressed osteon structure and altered soft tissue

arrangement, also wrinkles were observed along the cutting direction which explains

the overall shortening of specimen size (Figure 6.4h). This difference of bone tissue

morphology was attributed to the different mechanical force applied to the embedded

tissue from microtome blade during sectioning and bandsaw cutting followed by

grinder polishing.

Page 143: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 125

Figure 6.4 Specimen preparation and Goldner’s trichrome staining results of a tissue engineered sheep tibia with a 3 cm critical sized defect post mechanical testing. (a) Schematic illustration of sampling of tissue engineered sheep tibia bone. Note the tissue-engineered bone in the whole defect area and host bone on both opposing ends. (b) a 2 mm thick slab cut along the sagittal plane as depicted in the image via an Exakt cutting system. The specimen was then embedded in Technovit 9100 NEW® embedding media. The resin block was sectioned using a sledge microtome to generate (d) resin thin sections of ~10 μm, and (c) a resin ground section of ~50 μm with the Exakt cutting and grinding system. (d) The resin thin sections have undergone extensive shrinkage to a length of 4 cm, compared to (b) the original specimen before embedding (4.8 cm). Both resin ground and thin sections were stained with Goldner’s trichrome (e and f respectively). (g) High magnification images of resin ground section show Haversian canal (blood vessels in orange) and intact osteon structure. (h) High magnification images of thin section show Haversian canal (blood vessels in purple) and osteon structure at the identical location to that of the resin ground section. Noticeable alteration of Haversian

Page 144: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

126 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

canal structure including blood vessel location and ruffling in the bone matrix was observed compared to the resin ground section. Scale bar = 50 µm Resin microtomy creates tissue sections differently to ground sectioning. In ground

sectioning, as shown in Figure 6.2, resin embedded bone tissues were cut by a

diamond-edged saw and then carefully polished down to desired thickness by

sandpapers. The grinding process used a series of fine silicon carbide sandpapers

(P800 to P4000) with constant oscillation by the Exakt grinder to ensure a consistent

production of tissue sections without compression, folding or displacement tissue

components. The sledge microtome used a tungsten carbide blade with a plane-shaped

edge (profile D microtome knife) [222]. This type of microtome blade was extremely

hard and designed to cut hard materials such as resin to produce undecalcified bone

sections. However, the edge of the blade contributed to friction between the edge of

the blade and resin block [222]. The friction as the knife edge passed through the resin

block may have what caused compression of sections. The compression force can

cause plastic deformation of the embedded tissue and lead to shortened sections in the

cutting direction.

Cutting and distribution of TE bone specimens for paraffin and resin

histology

When investigating the bone regenerative capacity of a TE scaffold in vivo, it is

imperative to make the best use of limited bone tissue for the maximal amount of data.

I examine these bone explants in the order of: 1) micro-Computed Tomography (μCT)

analysis, 2) mechanical testing and lastly 3) histological analysis. To ensure a

comprehensive histomorphometry and immunohistochemistry (IHC) study of

specimens, the same tissue engineered bone was sampled for both paraffin and resin

histology. If we consider a 3-cm defect sheep tibia model for example (figure 6.5a),

the bone explants were cut as depicted in Figure 6.5b. The ~2 mm thick central bone

slabs around 4 cm in length were processed for resin embedding (Figure 6.5c) and the

remaining bone specimens were cut into 1.5 cm long for paraffin embedding (P1, P2,

and P3). Good tissue discrimination was obtained from resin ground sections stained

by Goldner’s trichrome and resin thin sections stained by von Kossa/MacNeal’s

tetrachrome stain (Figure 6.5d and e). The areas of mineralized bone indicated by

Goldner’s trichrome (green colour) closely match that shown by Von

Page 145: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 127

Kossa/MacNeal’s tetrachrome staining (black colour). The resin sections provide the

overview of the entire 3 cm bone defect site with ~0.5 cm host bone on both proximal

and distal end, which helps to understand the new bone integration with the host bone

at the scaffold/host bone interface. With the overview, the regenerated bone can be

quantified with OsteoMeasure system for detailed histomorphometric variables [223]

such as bone volume which are used to compare and validate the μCT analysis. Again,

the resin thin sections showed obvious compression along the cutting direction of

microtome blade, more significantly at defect sites with less regenerated bone (Figure

6.5e). The defect site with tissue engineered bone contributes to the compression as

only a small degree of compression was measured from the host bone area (by

comparing the black dashed lines to red dashed lines).

Compared to resin sections, the overall size of paraffin sections is limited by

standard embedding, sectioning and staining techniques. Even though oversized

specimens may be embedded in paraffin using custom-made large moulds, it is

challenging to obtain paraffin sections from these oversized paraffin blocks. Therefore

large specimens are regularly cut and divided into multiple parts to fit into the standard

embedding moulds for paraffin embedding. It is normally challenging to provide the

overview of such large specimens with a single paraffin section, but by placing

together the stained sections of all three parts (P1, P2, and P3) we can get an overview

of the whole specimen (Figure 6.5f). P1 shows the scaffold/proximal host bone

interface (Figure 6.5f-I), P3 shows the scaffold/distal host bone interface (Figure 6.5f-

III), while P2 shows the cross section of bone tissue within the scaffold (Figure 6.5f-

II).

Page 146: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

128 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Page 147: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 129

Figure 6.5 Specimen preparation and staining results of sheep tibiae with scaffolds implanted into 3 cm critical sized defects. (a) A schematic of a sheep tibia with a 3 cm scaffold in the defect. (b) Schematic illustration of how sheep tibia bone was cut and divided for both paraffin and resin embedding. (c) A 2 mm thick bone slab was cut from each tibia along the sagittal plane via an Exakt cutting system. The bone slabs were embedded in Technovit 9100 NEW® embedding media and produced into resin ground sections (~50 μm) by the Exakt cutting and grinding system, and into resin thin sections (~10 μm) by a sledge microtome. (d) The resin ground sections were stained with Goldner’s trichrome and (e) resin thin sections stained with Von Kossa/MacNeal’s tetrachrome staining. The areas of mineralized bone indicated by Goldner’s trichrome (green colour) closely match that shown by Von Kossa/MacNeal’s tetrachrome staining (black colour), however, the resin thin sections show obvious compression as a result of sledge microtome sectioning. The remaining bone specimens were cut into 3 slices (P1, P2, and P3) approximately 1.5 cm long for decalcification followed by paraffin embedding, and 5 μm sections were produced along cutting planes (I, II and III) as highlighted in (b). (f) Sections were stained with H&E staining and the images were put collated for an overview of bone tissue [224]

Staining of complex tissues (mouse paw)

On most occasions, bone fractures happen in a complex tissue environment often

associated with other tissue types such as cartilage, muscle, and skin. As a result, tissue

engineers are interested in the knowledge of structure and interfaces of such complex

tissues to enable better understanding of bone regeneration [225]. Mouse paws have

complex tissue types including bone, muscle, cartilage at joints and various connective

tissues. I used these samples to assess different sectioning approaches for complex

tissue to ascertain the most optimised approach for future bone TE histology strategies

(Figure 6.6). Goldner’s trichrome stain was used to discriminate these tissues on the

non-demineralized resin ground section (Figure 6.6a): bone (green), connective tissue

(orange), muscle (red) and joint cartilage (bright yellow). Similarly, the resin thin

section of the same paw had an identical colour differentiation between tissue types

(Figure 6.6b). However, the resin thin section showed distorted knuckle joint structure

(Figure 6.6b2) and shredded mineralized bone matrix (Figure 6.6b4). The connective

tissue and muscle was comparable in quality to that of resin ground section (Figure

6.6b3). The paraffin section had well preserved bone and joint morphology, but poor

hard/soft tissue interface, as shown by the clearly detached skin and muscle from the

bone. The paraffin section stained by Goldner’s trichrome staining did not provide

optimal tissue discrimination compared to H&E stained section (Figure 6.6d).

Page 148: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

130 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Figure 6.6 Comparison of mouse paws prepared as resin ground sections, resin thin sections and paraffin sections stained with Goldner’s trichrome stain. Comparative images of (1) an

Resin ground section

Resin thin section

Paraffin section

Paraffin section

Goldner’s trichrome

Goldner’s trichrome

Goldner’s trichrome

H&E

Page 149: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 131

overview, (2) knuckle joint, (3) muscle tissue and (4) metatarsal inter connective space show that the different sectioning techniques preserve tissue morphology differently. (a) The resin ground section of mouse paw embedded in methyl methacrylate (MMA) resin shows well preserved morphology of (a4) mineralized bone, (a2) joint cartilage and (a3) soft tissues including muscle, while (b) the sledge microtomed resin thin sections exhibit poor (b4) bone and (b2) joint morphology, although (b3) soft tissue is well preserved. (c) While the paraffin sections show well preserved (c4) bone and (c2) joint morphology, (c3) the soft tissue regions are poorly preserved. This figure highlights the versatile nature that ground sectioning has in the superior tissue morphology preservation of a complex hard/soft tissue environment as demonstrated in a mouse hind paw. Scale bar 1 = 2,000 µm, Scale bar 2-4 = 100 µm

Versatility

In our experience, the technique combination of resin ground sectioning and

Goldner’s trichrome staining provides the optimal bone tissue morphology

preservation and discrimination. These techniques are versatile and applicable to a

wide range of bone explant types. Figure 6.7a1 shows the sharp contrast overview

image of the mouse paw, and the tissue details of various types of a mouse paw

embedded in Technovit 9100 NEW® can be viewed with high magnification

microscope images (Figure 6.7a2 – a6), including digits, metatarsal joint, osteon

structure, muscle and blood vessels. Metallic implants are popular in bone tissue

engineering for their excellent mechanical properties, however they present a great

challenge for histological processing [6]. Our optimised techniques were applied to

bone defect containing a titanium implant to provide bone/implant contact information

without compromising the scaffold structure or tissue morphology (Figure 6.7b). The

porous titanium scaffolds were produced and implanted into sheep tibia by Seamus

Tredinnick et al. to investigate osteointegration of these scaffolds. The high

magnification images of tissue scaffold interface provide the information of bone

contact and osteointegration (Figure 6.7b2, b3 and b4).

Page 150: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

132 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Figure 6.7 Overview and high magnification images of a whole mouse hind paw and a sheep tibia/femur with porous titanium implant prepared by ground sectioning and stained with Goldner’s trichrome. Goldner’s Trichrome staining of the resin ground section produced high quality images, preserving the structure and providing high resolution detail. Both specimens were embedded in Technovit 9100 NEW® and ground sectioned to ~50 μm (b) with the Exakt cutting and grinding system. (a1) The morphology of the entire mouse paw (~27 μm) is shown, along with (a2) details of the nail, (a3) metatarsal joint, (a4) osteon detail, (a5) muscle structure, and (a6) blood vessel. (b1) The overall image shows the morphology of bone tissue integrated with the titanium implant. (b2 - b4) The high magnification images provide details of hard and soft tissue interactions around the metal implant. This figure highlights the high quality sections and supporting microscopic images obtainable from ground sectioning. Both overall tissue morphology and fine structures and details are preserved. Ground sectioning is the only histological technique capable of looking at metallic

Page 151: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 133

implant/hard and soft tissue interfaces, to date. Scale bars, a1 b1 = 2,000 µm, a2 = 50 µm, a3-a6 = 25 µm, b2 = 100 µm, b3-b4 = 100 µm

6.4 CONCLUSION OF STUDY 3

Study 3 aimed to develop the optimal endpoint assessment for the planned

implantation of PCL/SrBG scaffolds produced in previous studies. In this study, I

optimised and standardized essential techniques for histological processing of animal

native bone and tissue engineered bone explants. Once the PCL/SrBG scaffolds are

implanted in the future, they can be assessed using these optimised histological

methods developed in this study.

Study 3 was part of my PhD project and was inevitably constrained by time and

resources, preventing us from including the whole spectrum of animal species and

histological stains used in bone TE. Therefore, I standardized and compared

histological processing on identical bone specimens of four most frequently utilized

animal species in bone TE in vivo research. The optimised histological techniques have

been successfully applied on bone specimens with TE scaffolds implanted in projects

conducted by our colleagues and collaborators. I believe that the present study is a

major step towards optimal explant assessment by providing repeatable histological

processing techniques and clear results that researchers can use as a guide to their

experiment planning. As I have mentioned, different processing routes require

different specimen preparation that can be irreversible. For example, paraffin

embedding and sectioning require complete decalcification of bone specimen, which

eliminates the possibility of sharp discrimination using mineral specific stains.

In summary, the main lessons learned for tissue engineers and bone histologists

have been summarized in table 6.6 (different embedding media) and 6.7 (different

sectioning techniques for resin embedded specimens) and are summarized as follows.

Our staining results indicate that each embedding and sectioning technique has its

unique strength and disadvantages and should be employed based on a case-by-case

basis for a specific purpose. As the most routinely used histological technique, paraffin

embedding and sectioning is robust and easy to use. It has the advantages of high

throughput and efficiency, long storage life, and it provides well preserved bone

morphology and high quality H&E staining and IHC results. However, its limitations

Page 152: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

134 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

including small size, poor preservation of hard/soft tissue interfaces, the inability of

mineralized tissue discrimination due to complete mineral removal and slow

decalcification processes have rendered paraffin processing suboptimal for bone

explant assessment in tissue engineering. For researchers who are only interested in

the general morphology or have no access to specialised equipment for resin

embedding and sectioning, the paraffin approach is recommended for adequate results.

Resin embedding largely overcomes the limitations associated with paraffin approach

and the end results depend on sectioning techniques. The resin sledge microtome

sectioning is efficient in slide production and the best bone mineral discrimination by

von Kossa stain was observed on resin thin sections. However, the resin microtome

technique is found suboptimal at preserving the hard tissue morphology and hard/soft

tissue interface and a significant compression effect is observed. This technique is also

unreliable in cutting mature cortical bone specimens. On the other hand, the resin

ground sectioning technique provides the best bone tissue/explant morphology

especially in complex and large sized specimens. Goldner’s trichrome stain offers a

sharp contrast to tissue types by colour and maximizes the data output of a single

section. The combination of ground sectioning and Goldner’s trichrome techniques

brings the user the best overview of morphology with good cellular details. Little

difference in morphological staining results was observed between MMA and

Technovit 9100 NEW® resin ground sections, and immunolabelling was successful

only on Technovit 9100 NEW® ground sections. Two noticeable disadvantages of

ground sectioning techniques are the relatively large amount of specimen consumption

per section (owing to the grinding phase) and requirement of specialized equipment

and resources.

Table 6.6 Summary of the advantages and disadvantages of commonly used media for histology embedding: paraffin, MMA resin and Technovit 9100 resin.

Embedding

media

Advantages Disadvantages

Paraffin • robust and reliable, efficient in

tissue section production which is

• poor hard/soft tissue interface

• no preservation of minerals

Page 153: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 135

critical for TE implants small in

size

• paraffin sections are ideal for H&E

staining and IHC which provide

high quality cellular details, matrix

morphology, specific markers of

regenerated bone on TE scaffolds

• automated processing, staining and

slides scanning (optional)

• relatively cheap to process

• limited specimen size

• not suitable for metal implants

• Collapsing or damage of

scaffold explant observed due

to melting or dissolving of

scaffolds structures

MMA resin • preserves mineral contents of

regenerated bone on scaffold

implants

• allows bone specific stains that

provide differentiation of hard/soft

tissues, which is crucial for the

assessment of osteogenic capacity

of scaffold implants

• allows oversized specimens to be

processed with integrity. This is

especially important for scaffold

implantation experiments in large

animal models (such as sheep and

pig)

• time consuming and laborious

• low level of automation

• damage to antigens thus IHC

not possible

Technovit

9100 resin

• preserves mineral contents of

regenerated bone on scaffold

implants

• allows bone specific stains that

provide differentiation of hard/soft

• time consuming and laborious

• low level of automation

• relatively expensive

Page 154: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

136 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

tissues, which is crucial for the

assessment of osteogenic capacity

of scaffold implants

• allows oversized specimens to be

processed with integrity. This is

especially important for scaffold

implantation experiments in large

animal models (such as sheep and

pig)

• preserves the antigens and enables

IHC analysis

Table 6.7 Summary of the advantages and disadvantages of commonly used sectioning techniques for resin embedded specimens

Sectioning

technique

Advantages Disadvantages

Sledge

microtome • efficient in section

production

• shrinking effect

• poor preservation of hard tissue

morphology

• can alter scaffold implant

morphology leading to

complications for quantitative

analysis

• time consuming

Ground

sectioning • best in tissue morphology

(hard and soft) preservation

• consumes relatively large

volumes of the specimen

(specimen depletion), which is

not desirable for scaffold implant

small in size

Page 155: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 137

• scaffold morphology

maintained as pre-implant

conditions

• can cut extremely hard

materials (specimens with

metal implants)

• very time consuming

The results of the present study will serve as a standard for testing the

performance of histological techniques for bone and as a guide to researchers for their

bone histological analysis.

6.5 SUPPLEMENTARY FIGURES

Page 156: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

138 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Supplementary Figure 6.1 Comprehensive comparison of H & E staining on bone sections prepared by different embedding techniques of all four animal species. The resin ground sections are ~27 µm in thickness, and paraffin sections are 5 µm in thickness. Cell nuclei are stained dark purple/blue, while cytoplasm is stained pink. Inserts show overall images of the entire stained tissue. For the ease of comparison, the images are categorized by both animal species: (a,b,c) sheep tibiae, (d,e,f) pig fibulae, (g,h,i) mouse paws, and (j,k,l) rat leg boned; and types of embedding media: (a,d,g,j) MMA, (b,e,h,k) Technovit 9100 New®, and (c,f,i,l) paraffin sections. Scale bar = 25 μm

Page 157: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants 139

Supplementary Figure 6.2 Comprehensive comparison of Von Kossa/MacNeal’s tetrachrome staining on bone sections prepared by different embedding techniques of all four animal species. The resin ground sections are ~27 µm in thickness, and paraffin sections are 5 µm in thickness. Mineralized bone tissue stains dark brown/black while soft tissue stains light blue and cell nuclei stain dark blue. Inserts show overall images of the entire stained tissue. For the ease of comparison, the images are categorized by both animal species: (a,b,c) sheep tibiae, (d,e,f) pig fibulae, (g,h,i) mouse paws, and (j,k,l) rat leg boned; and types of embedding media: (a,d,g,j) MMA, (b,e,h,k) Technovit 9100 New®, and (c,f,i,l) paraffin sections. Scale bar = 25 µm

Page 158: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

140 Chapter 6: (Study 3) Development of optimised histological processes for analysis of large and complex bone and implants

Supplementary Figure 6.3 Comprehensive comparison of Immunohistochemistry on sections of bone embedded in Technovit 9100 New® and paraffin for all four animal species. The resin ground sections are ~27 µm in thickness, and paraffin sections are 5 µm in thickness. (a1-a8) Collagen type I (2μg/ml polyclonal rabbit), (b1-b8) osteocalcin (2μg/ml polyclonal rabbit), (c1-c8) Von Willebrand factor (vWF) (ready-to-use, polyclonal rabbit anti human). (d1-d8) Negative controls for immunohistochemistry using rabbit primary antibody isotype control (0.5μg/ml)

Page 159: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

141

Chapter 7: Conclusions

7.1 RESEARCH SUMMARY

Nationally and globally, musculoskeletal disorders place a significant burden on

the healthcare system and overall economy, and in Australia, they account for $15

billion annual direct health expenditure [4,42]. The clinical treatment for these

conditions relies on bone grafting: autografting (donor tissue from the patient) and

allografting (donor tissue from another person). Despite these treatment options all

exhibiting bone defect healing capacity, they also have shown prominent

disadvantages [28]. The need for better treatment has driven researchers to develop

bone graft substitute and led to the emergence of tissue engineering field [31].

Scaffold-based tissue engineering approach investigates the use of biomaterials

(polymers, bioceramics etc.) as potential bone graft substitutes, potentially

overcoming many of the issues associated with autologous and allogeneic bone

grafting. In general, the development of scaffolds involves three stages (Figure 1.1),

which starts with scaffold fabrication and characterization (stage 1), followed by in

vitro cell assays (stage 2) and in vivo implantation with rigorous tissue explant

assessment (stage 3) to evaluate the efficacy and viability of scaffolds. Histology is

considered as one of the essential explants analytical methods for TE scaffolds.

In this PhD project, I hypothesized that by incorporating strontium-substituted

bioactive glass (SrBG) particles into polycaprolactone (PCL) I could increase the

bioactivity of PCL and produce porous scaffolds. To develop the composite scaffold,

I started with stage 1 – scaffold fabrication via melt-electrospinning technique and

characterization, following the successful production of PCL/SrBG (10 wt%) I worked

on stage 2 of in vitro assessment, to find out the osteoblast-like cells attachment,

proliferation, and osteoblast differentiation on the composite scaffolds compared to

PCL only control scaffolds; and in parallel, I optimised histological techniques for

bone specimen examination for the optimal assessment of intended in vivo

implantation of these composite scaffolds.

Page 160: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

142

7.2 SUMMARY OF CHAPTER 4 (STUDY 1)

Chapter title: Fabrication and in vitro investigation of PCL, 10 wt% PCL/SrBG

electrospun scaffolds for bone regeneration

Chapter 4 demonstrated for the first time the development of a PCL/SrBG

composite scaffold incorporating 10% (by weight) of SrBG particles into PCL bulk,

produced by the technique of melt-electrospinning. The PCL/SrBG composite

scaffolds were reproducibly manufactured with an interconnected porous structure.

MicroCT analysis showed the homogeneous distribution of SrBG particles throughout

the scaffold matrix. In vitro bioactivity of the composite scaffolds was investigated

with ICP-OES and EDX techniques. Quantitative analysis of ions dissolution in cell

culture media and evidence of CaP layer precipitation on scaffold surface indicated

similar bioactivity to that of bare SrBG particles.

MC3T3 cells were cultured on these scaffolds in two groups: normal cell culture

media and osteogenic media (normal culture media supplemented with osteogenic

factors). A series of in vitro assays were carried out. The PCL/SrBG scaffolds were

demonstrated to be non-cytotoxic in vitro. Ions present in the SrBG component were

shown to dissolve into cell culture media and promoted precipitation of a calcium

phosphate layer on the scaffold surface which in turn led to noticeably enhanced

alkaline phosphatase (ALP) activity in MC3T3-E1 cells compared to PCL only

scaffolds. Furthermore, up-regulation of ALP and OCN genes was also observed.

These results indicated PCL/SrBG composite scaffolds could enhance osteoblasts

differentiation in vitro. The composite scaffolds were also found to enhance collagen

deposition. These results suggested that melt-electrospun PCL/SrBG composite

scaffolds could noticeably enhance bone differentiation compared to PCL ones, and

the PCL/SrBG scaffolds showed potential to become effective bone graft substitutes.

However, the PCL/SrBG scaffolds did not show such effect when exposed to normal

cell culture media mainly due to insufficient bioactivity. Therefore, it was imperative

for me to increase the SrBG weight percentage in PCL to render an enhanced

bioactivity and thus bone differentiation of the PCL/SrBG scaffolds, which led to the

study 2.

Page 161: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

143

7.3 SUMMARY OF CHAPTER 5 (STUDY 2)

Chapter title: Developing 50 wt% Strontium-substituted bioactive glass and

Polycaprolactone composite scaffolds for bone repair via hybrid electrospinning in a

direct writing mode

Following the research on 10% PCL/SrBG scaffolds in study 1, I actively

investigated the way of increasing the weight percentage of SrBG particles loaded in

the composite scaffolds in order to further enhance their bioactivity. In chapter 5, I

successfully fabricated PCL/SrBG scaffolds containing 50 wt% of SrBG with

controlled porosity and structure via melt-electrospinning in a direct writing mode. In

order to achieve this, I optimised the whole process including SrBG particle sizing,

PCL/SrBG composite preparation and the electrospinning technique. Firstly, the SrBG

particle size was reduced from over 100 μm to < 6 μm to minimise the risk of needle

blockage, which also led to increased surface area of these particles. The solvent

precipitation technique was adopted for the 50 wt% PCL/SrBG composite preparation

to ensure a homogeneous distribution of particles within the PCL matrix.

The production of the composite scaffolds was very challenging and in respond

I developed a novel hybrid melt-electrospinning system for the PCL/SrBG scaffold

production. This hybrid technique facilitated composite fibre continuity and fibre jets

stability for the precise control of electrospun fibres deposition. Compared to

PCL/SrBG (10 wt%) scaffold in study 1, the PCL/SrBG (50 wt%) scaffolds showed

greatly enhanced in vitro bioactivity indicated by the faster formation of CaP layer

only after 3 hours in serum-free media. More importantly, the concentration of Sr and

Si ions released from the PCL/SrBG (50 wt%) scaffolds also increased and the peak

values were comparable to that of bare SrBG particles. As a control, PCL only

scaffolds did not show CaP formation or Sr and Si ion dissolution.

In vitro tests were performed on these PCL/SrBG (50 wt%) scaffolds to evaluate

the osteogenic capacity of these scaffolds. Same as in study 1, MC3T3 cells were

cultured on PCL/SrBG and PCL scaffolds in both osteogenic media and growth control

media. A series of in vitro assays were carried out for the assessment of cell attachment,

proliferation and osteoblastic differentiation in all experimental conditions. Based on

LIVE/DEAD assay, SEM, CLSM and MTT assay results, the PCL/SrBG (50 wt%)

scaffolds were not cytotoxic and had the ability to support cell attachment and

proliferation. Notably, the significant increase of ALP activity of cells cultured on

Page 162: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

144

PCL/SrBG scaffolds in growth media compared to their counterpart on PCL scaffolds

indicated the PCL/SrBG were able to induce osteoblast differentiation without

osteogenic supplement in the media, which made the composite scaffolds osteogenic

in vitro. Subsequent gene expression analysis showed upregulation of ALP and OPN

genes confirmed the osteogenic capacity of these PCL/SrBG (50 wt%) scaffolds.

Previous studies have shown that composite scaffolds were implanted for in vivo

investigation without in vitro confirmation of the osteogenic capacity of these

scaffolds [160]. Other studies investigate the osteogenic capacity of the scaffolds using

in vitro cell investigation prior to in vivo implantation using only osteogenic media

[75]. In this study, I made a step further and investigated the osteogenic potentials of

the PCL/SrBG composite scaffolds without osteogenic media in vitro, so we could

understand the real efficacy of these scaffolds to enhance osteogenesis. The findings

of this study indicated that these PCL/SrBG scaffolds with increased SrBG loading

were suitable for next stage of in vivo implantation.

7.4 SUMMARY OF CHAPTER 6 (STUDY 3)

Chapter title: Development of optimised histological processes for analysis of

large and complex bone and implants

As an essential endpoint tool for TE scaffold ex vivo assessment, histology has

become a key stage of TE scaffold development. Therefore in parallel to the

PCL/SrBG composite scaffold fabrication and in vitro evaluation in study 1 and 2, I

conducted study 3 to develop the optimised histological techniques for the ex vivo

assessment of the composite scaffolds developed in this project. To include all the

animal species in which the PCL/SrBG scaffolds were likely to be implanted, I

designed a comprehensive study to histologically process four most frequently utilized

animal species in bone tissue engineering in vivo research: sheep, pig, rat, and mouse.

In this study, I standardized and compared histological processing on identical bone

specimens embedded in paraffin, MMA resin, and Technovit 9100 NEW® resin.

Paraffin sections were obtained by microtomy and resin sections by sledge microtome

and ground sectioning technique. Common histological stains and bone specific stains

including H&E, Von Kossa, Goldner’s trichrome staining and IHC were performed on

all these sections. The stained sections were compared across all animal species to

reveal the difference between different processing techniques and to identify the

optimal histological techniques for bone analysis. The histological techniques

Page 163: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

145

standardized in this study were applicable to mineralized tissues such as bone and

teeth, as well as regenerated bone via TE scaffold implantation. With the optimised

histological analysing techniques, we were poised to analyse the PCL/SrBG scaffolds

once they were explanted. Even though the PCL/SrBG composite scaffolds could not

be implanted due to time constrains of my PhD, I believe that the present study was a

major step towards optimal explant assessment by providing repeatable histological

processing techniques and detailed corresponding results that researchers can use as a

guide in their experiment planning.

7.5 LIMITATIONS AND RECOMMENDATION FOR FUTURE WORK

The PCL/SrBG (10 wt%) composites were fabricated into scaffolds via melt-

electrospinning technique for the first time and showed enhanced osteogenic capacity.

Further effort was made to fabricate scaffolds with increased SrBG filler phase and

resulted in PCL/SrBG (50 wt%) scaffolds via a novel hybrid melt-electrospinning

technique. The PCL/SrBG (50 wt%) scaffolds showed greatly enhanced in vitro

bioactivity compared to the PCL/SrBG (10 wt%) scaffolds. These PCL/SrBG scaffolds

were found osteogenic in vitro. However, more work needs to be done to optimise the

scaffold design and determine the osteoinductive capacity of these scaffolds through

in vivo implantation. The limitations and recommended future work of this PhD project

are discussed in the following sections.

7.5.1 Composite scaffold design

In order to obtain straight electrospun fibres, a short needle tip to collector

distance was used in study 2 during the 50% PCL/SrBG scaffolds fabrication, resulting

in larger fibre diameters compared to PCL ones. A series of in vitro assessment can be

done to determine the optimum fibre diameter. In situations where straight fibres are

not imperative, this collection distance may be increased for reduced fibre diameters.

In this project, I reduced the SrBG particle size by grinding for improved fibre

continuity but I could not modify the irregular shape of the particles and their sizes

were not uniform. Ideally, spherical nano-sized SrBG particles, which provide a much

higher surface area to volume ratio compared to the current micron-sized particles,

would be used in future studies to further improve the electrospun fibre production. As

we know, the particle size, size distribution and particle filler percentage within

composite play an important role in the mechanical properties of the PCL/SrBG

Page 164: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

146

composite scaffolds. The enhanced 50 wt% SrBG filler phase in the PCL/SrBG

scaffolds developed in this PhD project was a trade-off of bioactivity (resulted from

high SrBG loading) and the mechanical integrity of the scaffolds. Further increasing

the SrBG filler phase may boost the bioactivity of PCL/SrBG composite, however, the

increased amount of SrBG particles will probably make the PCL/SrBG scaffold brittle.

In this PhD project, the PCL/SrBG (50 wt%) scaffolds were fabricated in a 0/90

degrees cross-hatch structure with a fibre space of 1mm. With the motorized stage and

aid of G code, other scaffold lay-down pattern can be explored to obtain different

scaffold structure such as 0/60/120 degree lay-down pattern. Shorter fibre spacing can

be adopted for reduced pore sizes which would facilitate bridging of scaffold pores by

seeded cells. Additionally, a rotating mandrel collector can be used to make porous

tubes out of the PCL/SrBG composites. During the hybrid electrospinning process, the

impact of SrBG incubation in organic solvents can be further investigated using bare

SrBG particles. The SrBG particles will be weighed prior and after immersion in

chloroform for a pre-determined time at 60 °C to check if there will be any possible

dissolution or degradation. The incubated SrBG particles can also be analysed using

ICP-MS technique, and the results of elemental concentration will be compared with

the non-immersed SrBG. These studies will help to determine whether the

characteristics of SrBG would alter during the scaffold fabrication process.

Furthermore, to understand the interaction of elemental concentrations (Ca, P,

Si and Sr) and cell activities, both osteogenic and control cell culture media in future

in vitro studies should be collected and analysed with ICP-MS technique. More

specifically, each scaffold for cell culture should be weighed and time interval and

volume of media change in each tissue culture plate well should be predetermined.

Triplicates of media at each time point should be collected and their ion concentration

measured.

7.5.2 In vivo investigation of composite scaffolds

Before these composite scaffolds may be used as bone graft substitutes in clinical

treatment for bone defects, they must be rigorously tested under in vivo conditions.

The composite scaffolds are recommended to be implanted into rat cranial defects for

the assessment of their osteogenic capacity. Post-operative assessment for bone

explants includes microcomputed tomography (μCT), mechanical testing and bone

Page 165: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

147

histology, all of which are well optimised. The detailed plan is described in the

following sections.

Critical defect production and scaffold implantation via animal surgery

Twelve skeletally mature male Wistar rats will be used in three groups: 1)

PCL/SrBG scaffolds group; 2) PCL scaffolds and 3) empty control group. All rats will

be operated under general anesthesia. Two bone defects of 5mm in diameter will be

created in full thickness per calvaria. The animals will be sacrificed, by CO2 inhalation,

12 weeks post surgery and bone explants will be collected for further analysis using

techniques similar to previous study described by Berner et al. (2013) [107].

Subcutaneous injections of fluorochrome labelling (Tetracycline hydrochloride,

Alizarin complexone and calcein) will be administered at defined time points after the

surgery to identify bone mineral deposition rate [226].

μCT analysis

A Micro-CT 40 scanner (SCANCO Medical, Bassersdorf, Switzerland) will be

used to quantify the mineralization within the defects, presented as bone volume in

mm3.

Mechanical testing

An Instron Micro tester 5848 will be used to perform non-destructive micro-

compression tests on the calvaria defects to evaluate the mechanical stiffness of

regenerated bone in the defect.

Histological analysis

Following mechanical testing, the bone explant specimens will be fixed in 4%

paraformaldehyde in PBS for 48 h. These specimens will be divided for both paraffin

and resin embedding with five defects per group. For paraffin embedding, the

specimens will be decalcified in 10% Ethylenediaminetetraacetic acid (EDTA) for 3-

4 weeks. The 5-μm paraffin sections produced by microtome will be used for

histomorphometry and immunohistochemistry (IHC) analysis. The specimens

intended for resin embedding will be dehydrated and embedded in Technovit 9100

NEW®. Resin sections will be produced by a sledge microtome (Polycut-S, Reichert-

Jung, International Medical Equipment, USA) at the thickness of 6 μm. The resin

sections will be stained with Goldner’s trichrome and von Kossa/MacNeal’s

tetrachrome stain to identify new bone formation.

Page 166: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

148

7.6 CONCLUDING REMARKS

With the increasing demand for autografts in the clinical setting, tissue engineers

are developing TE scaffolds as promising substitutes for bone graft materials.

Developing bioactive materials and improving fabrication techniques for scaffold

production are the research focus for viable scaffolds in bone TE. In the attempt to

improve the bioactivity of polymers, the use of polymer/BG composite biomaterial has

shown superior osteoconductive and even osteoinductive properties to single-

component polymers. The bioactive composite scaffolds have shown potential to be

used for the purpose of bone repair without cells or growth factors.

There has been a paradigm shift towards produce TE scaffolds using additive

manufacturing (AM) techniques to fabricate scaffolds with desired structure and

porosity. The AM scaffolds fabrication techniques or also known as 3D printing

produce scaffolds with layer-by-layer fibre deposition, and the 3D anatomically

precise implants are promising as future personalised medicine. Melt-extrusion based

techniques such as FDM have been used to produce porous composite scaffolds with

controllable size, shape and porosity. FDM scaffolds normally consist of fibres around

500 μm in diameter. In comparison, melt-electrospinning technique can produce

scaffolds fibres down to micron level which lead to better structural resolution and

higher surface area to volume ratio than the FDM scaffolds. The recent advances in

melt-electrospinning have enabled researchers to fabricate scaffolds with desired

structure and porosity. However, there are limited publications on composite scaffold

fabrication with this technique due to the difficulties associated with the change of

viscoelastic properties of polymers when inorganic phase was incorporated.

This PhD project aims to bridge this knowledge gap by advancing the melt-

electrospinning technique for the production of polymer/BG composite scaffolds. In

this PhD project, the PCL/SrBG composite scaffold with up to 50 wt% SrBG loading

was successfully fabricated. These composite scaffolds were bioactive and osteogenic

in vitro, which provided more options for the future off-the-shelf and patient specific

solutions for bone defect treatment. Another contribution of this project is the

advancing of the melt-electrospinning technique for the production of composite

scaffolds with aligned fibre deposition. This novel technique opens up the possibility

to fabricate PCL/SrBG scaffolds with different SrBG loading, or even the production

of PCL scaffolds incorporated with a whole different array of inorganic materials for

Page 167: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

149

specific purposes. Lastly, in pursuance of preparing optimal histological assessment

tools for the planned in vivo implantation experiments, this PhD project also provided

recommendations, general guide and optimised operating protocols for the endpoint

assessment of bone tissues/scaffold explant for researchers in bone TE field.

In conclusion, this PhD project contributes to the overall tissue engineering

research towards effective and patient-specific treatment of tissue loss of human body

using synthetic scaffolds. However, being the first to fabricate the melt-electrospun

PCL/SrBG scaffolds has made the body of work in this PhD project an indispensable

link in understanding of future cell-free treatment for bone defects with fully synthetic

bioactive scaffolds.

Page 168: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J
Page 169: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 151

References

[1] M.J. Chehade, A. Bachorski, Development of the Australian Core Competencies in Musculoskeletal Basic and Clinical Science project - phase 1, Med. J. Aust. 189 (2008) 162–165. http://www.ncbi.nlm.nih.gov/pubmed/18673105.

[2] (Aihw), Australia’s Health 2014., 2014. doi:Cat. no. AUS 178. [3] Arthritis and Osteoporosis Victoria, A Problem worth solving - The rising cost

of musculoskeletal conditions in Australia, (2013). [4] Health-care expenditure on arthritis and other musculoskeletal conditions

2008–09, 2008. [5] B.T. King, How great is the burden of musculoskeletal disease ?, (2008). [6] Y.H. An, K.L. Martin, Handbook of histology methods for bone and cartilage,

Humana Press, Totowa, N.J. ; [Great Britain], 2003. [7] R. Drury, Theory and Practice of Histological Techniques, J. Clin. Pathol. 36

(1983) 609. doi:10.1136/jcp.36.5.609-d. [8] E. Willbold, F. Witte, Histology and research at the hard tissue-implant

interface using Technovit 9100 New embedding technique., Acta Biomater. 6 (2010) 4447–55. doi:10.1016/j.actbio.2010.06.022.

[9] G. Schmidmaier, R. Capanna, B. Wildemann, T. Beque, D. Lowenberg, Bone morphogenetic proteins in critical-size bone defects: what are the options?, Injury. 40 Suppl 3 (2009) S39-43. doi:10.1016/S0020-1383(09)70010-5.

[10] K.S. Cahill, J.H. Chi, A. Day, E.B. Claus, Prevalence, complications, and hospital charges associated with use of bone-morphogenetic proteins in spinal fusion procedures., JAMA. 302 (2009) 58–66. doi:10.1001/jama.2009.956.

[11] E.J. Carragee, E.L. Hurwitz, B.K. Weiner, A critical review of recombinant human bone morphogenetic protein-2 trials in spinal surgery: emerging safety concerns and lessons learned., Spine J. 11 (2011) 471–91. doi:10.1016/j.spinee.2011.04.023.

[12] C. Mota, D. Puppi, F. Chiellini, E. Chiellini, Additive manufacturing techniques for the production of tissue engineering constructs, J. Tissue Eng. Regen. Med. (2012). doi:10.1002/term.1635.

[13] S. V Murphy, A. Atala, 3D bioprinting of tissues and organs., Nat. Biotechnol. 32 (2014) 773–785. doi:10.1038/nbt.2958.

[14] P.S.P. Poh, D.W. Hutmacher, M.M. Stevens, M. a Woodruff, Corrigendum: Fabrication and in vitro characterization of bioactive glass composite scaffolds for bone regeneration (2013 Biofabrication 5 045005), Biofabrication. 6 (2014) 29501. doi:10.1088/1758-5082/6/2/029501.

[15] B. Rolfe, The Fibrotic Response to Implanted Biomaterials: Implications for Tissue Engineering, Regen. Med. Tissue Eng. - Cells Biomater. (2011) 552–568. doi:10.5772/21790.

[16] J.R. Jones, Review of bioactive glass: From Hench to hybrids, Acta Biomater.

Page 170: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

152 References

9 (2013) 4457–4486. doi:10.1016/j.actbio.2012.08.023. [17] J. Yao, S. Radin, P. S. Leboy, P. Ducheyne, The effect of bioactive glass

content on synthesis and bioactivity of composite poly (lactic-co-glycolic acid)/bioactive glass substrate for tissue engineering, Biomaterials. 26 (2005) 1935–1943. doi:10.1016/j.biomaterials.2004.06.027.

[18] S. Weiner, H.D. Wagner, THE MATERIAL BONE: Structure-Mechanical Function Relations, Annu. Rev. Mater. Sci. 28 (1998) 271–298. doi:10.1146/annurev.matsci.28.1.271.

[19] T.J. Webster, E.S. Ahn, Nanostructured biomaterials for tissue engineering bone, Adv. Biochem. Eng. Biotechnol. 103 (2006) 275–308. doi:10.1007/10_021.

[20] M.J. Olszta, X. Cheng, S.S. Jee, R. Kumar, Y.-Y. Kim, M.J. Kaufman, E.P. Douglas, L.B. Gower, Bone structure and formation: A new perspective, Mater. Sci. Eng. R Reports. 58 (2007) 77–116. doi:10.1016/j.mser.2007.05.001.

[21] A. Nather, Bone grafts and bone substitutes : basic science and clinical applications, World Scientific, Hackensack, N.J., 2005.

[22] F. Barrère, T.A. Mahmood, K. de Groot, C.A. van Blitterswijk, Advanced biomaterials for skeletal tissue regeneration: Instructive and smart functions, Mater. Sci. Eng. R Reports. 59 (2008) 38–71. doi:10.1016/j.mser.2007.12.001.

[23] J.R. Porter, T.T. Ruckh, K.C. Popat, Bone tissue engineering: a review in bone biomimetics and drug delivery strategies, Biotechnol. Prog. 25 (2009) 1539–1560. doi:10.1002/btpr.246.

[24] R.A.D. Carano, E.H. Filvaroff, Angiogenesis and bone repair, Drug Discov. Today. 8 (2003) 980–989. doi:10.1016/s1359-6446(03)02866-6.

[25] R. Dimitriou, E. Jones, D. McGonagle, P. V Giannoudis, Bone regeneration: current concepts and future directions., BMC Med. 9 (2011) 66. doi:10.1186/1741-7015-9-66.

[26] J.E. Schroeder, R. Mosheiff, Tissue engineering approaches for bone repair: Concepts and evidence, Injury. 42 (2011) 609–613. doi:10.1016/j.injury.2011.03.029.

[27] a S. Brydone, D. Meek, S. Maclaine, Bone grafting, orthopaedic biomaterials, and the clinical need for bone engineering., Proc. Inst. Mech. Eng. Part H J. Eng. Med. 224 (2010) 1329–1343. doi:10.1243/09544119JEIM770.

[28] M.K. Sen, T. Miclau, Autologous iliac crest bone graft: Should it still be the gold standard for treating nonunions?, Injury. 38 (2007) S75–S80. doi:10.1016/j.injury.2007.02.012.

[29] C.J. Damien, J.R. Parsons, Bone graft and bone graft substitutes: a review of current technology and applications, J. Appl. Biomater. 2 (1991) 187–208. doi:10.1002/jab.770020307.

[30] D.L. Wheeler, W.F. Enneking, Allograft bone decreases in strength in vivo over time., Clin. Orthop. Relat. Res. (2005) 36–42. doi:10.1097/01.blo.0000165850.58583.50.

[31] J. Viola, B. Lal, O. Grad, The emergence of tissue engineering as a research field, Natl. Sci. Found. Arlington, Virginia, USA. (2003).

Page 171: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 153

[32] R. Langer, J.P. Vacanti, Tissue engineering, Science (80-. ). 260 (1993) 920–926. http://www.ncbi.nlm.nih.gov/pubmed/8493529.

[33] R. Langer, Perspectives and Challenges in Tissue Engineering and Regenerative Medicine, Adv. Mater. 21 (2009) 3235–3236. doi:10.1002/adma.200902589.

[34] A. Sherwin, R. Langer, D. Sc, Progress in the Tissue Engineering and Stem Cell Industry “‘ Are we there yet ?,’” 18 (2012). doi:10.1089/ten.teb.2011.0553.

[35] Y. Huang, M.C. Leu, J. Mazumder, A. Donmez, Additive Manufacturing: Current State, Future Potential, Gaps and Needs, and Recommendations, J. Manuf. Sci. Eng. 137 (2014) 1–10. doi:10.1115/1.4028725.

[36] G.H. Wu, S.H. Hsu, Review: Polymeric-based 3D printing for tissue engineering, J. Med. Biol. Eng. 35 (2015) 285–292. doi:10.1007/s40846-015-0038-3.

[37] I. Zein, D.W. Hutmacher, K.C. Tan, S.H. Teoh, Fused deposition modeling of novel scaffold architectures for tissue engineering applications., Biomaterials. 23 (2002) 1169–85. http://www.ncbi.nlm.nih.gov/pubmed/11791921.

[38] F.P.W. Melchels, K. Bertoldi, R. Gabbrielli, A.H. Velders, J. Feijen, D.W. Grijpma, Mathematically defined tissue engineering scaffold architectures prepared by stereolithography, Biomaterials. 31 (2010) 6909–6916. doi:10.1016/j.biomaterials.2010.05.068.

[39] J.M. Williams, A. Adewunmi, R.M. Schek, C.L. Flanagan, P.H. Krebsbach, S.E. Feinberg, S.J. Hollister, S. Das, Bone tissue engineering using polycaprolactone scaffolds fabricated via selective laser sintering, Biomaterials. 26 (2005) 4817–4827. doi:10.1016/j.biomaterials.2004.11.057.

[40] H.J. Yen, C.S. Tseng, S.H. Hsu, C.L. Tsai, Evaluation of chondrocyte growth in the highly porous scaffolds made by fused deposition manufacturing (FDM) filled with type II collagen, Biomed. Microdevices. 11 (2009) 615–624. doi:10.1007/s10544-008-9271-7.

[41] T.M. Seck, F.P.W. Melchels, J. Feijen, D.W. Grijpma, Designed biodegradable hydrogel structures prepared by stereolithography using poly(ethylene glycol)/poly(d,l-lactide)-based resins, J. Control. Release. 148 (2010) 34–41. doi:10.1016/j.jconrel.2010.07.111.

[42] M.J. Chehade, A. Bachorski, Development of the Australian Core Competencies in Musculoskeletal Basic and Clinical Science project - phase 1, Med. J. Aust. 189 (2008) 162–165. http://www.ncbi.nlm.nih.gov/pubmed/18673105.

[43] D.W. Hutmacher, S. Cool, Concepts of scaffold-based tissue engineering--the rationale to use solid free-form fabrication techniques, J. Cell. Mol. Med. 11 (2007) 654–669. doi:10.1111/j.1582-4934.2007.00078.x.

[44] A. Kolk, J. Handschel, W. Drescher, D. Rothamel, F. Kloss, M. Blessmann, M. Heiland, K.-D.D. Wolff, R. Smeets, Current trends and future perspectives of bone substitute materials - from space holders to innovative biomaterials., J. Craniomaxillofac. Surg. 40 (2012) 706–18. doi:10.1016/j.jcms.2012.01.002.

[45] M.A. Woodruff, D.W. Hutmacher, The return of a forgotten polymer—Polycaprolactone in the 21st century, Prog. Polym. Sci. 35 (2010) 1217–1256.

Page 172: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

154 References

doi:10.1016/j.progpolymsci.2010.04.002. [46] H.-H. Lee, H.-S. Yu, J.-H. Jang, H.-W. Kim, Bioactivity improvement of

poly(ε-caprolactone) membrane with the addition of nanofibrous bioactive glass, Acta Biomater. 4 (2008) 622–629. doi:10.1016/j.actbio.2007.10.013.

[47] J.S. Park, J.-M. Kim, S.J. Lee, S.G. Lee, Y.-K. Jeong, S.E. Kim, S.C. Lee, Surface hydrolysis of fibrous poly(ε-caprolactone) scaffolds for enhanced osteoblast adhesion and proliferation, Macromol. Res. 15 (2007) 424–429. doi:10.1007/BF03218809.

[48] M.N. Rahaman, D.E. Day, B.S. Bal, Q. Fu, S.B. Jung, L.F. Bonewald, A.P. Tomsia, Bioactive glass in tissue engineering, Acta Biomater. 7 (2011) 2355–2373. doi:10.1016/j.actbio.2011.03.016.

[49] A. Hoppe, N.S. Guldal, A.R. Boccaccini, A review of the biological response to ionic dissolution products from bioactive glasses and glass-ceramics, Biomaterials. 32 (2011) 2757–2774. doi:10.1016/j.biomaterials.2011.01.004.

[50] K. Rezwan, Q.Z. Chen, J.J. Blaker, A.R. Boccaccini, Biodegradable and bioactive porous polymer/inorganic composite scaffolds for bone tissue engineering, Biomaterials. 27 (2006) 3413–3431. doi:10.1016/j.biomaterials.2006.01.039.

[51] L.L. Hench, Bioceramics: From Concept to Clinic, J. Am. Ceram. Soc. 74 (1991) 1487–1510. doi:10.1111/j.1151-2916.1991.tb07132.x.

[52] C. Wu, J. Chang, A review of bioactive silicate ceramics., Biomed. Mater. 8 (2013) 32001. doi:10.1088/1748-6041/8/3/032001.

[53] L.L. Hench, H. a Paschall, Direct chemical bond of bioactive glass-ceramic materials to bone and muscle., J. Biomed. Mater. Res. 7 (1973) 25–42. doi:10.1002/jbm.820070304.

[54] L.L. Hench, R.J. Splinter, W.C. Allen, T.K. Greenlee, Bonding mechanisms at the interface of ceramic prosthetic materials, J. Biomed. Mater. Res. 5 (1971) 117–141. doi:10.1002/jbm.820050611.

[55] P.J. Meunier, C. Roux, S. Ortolani, M. Diaz-Curiel, J. Compston, P. Marquis, C. Cormier, G. Isaia, J. Badurski, J.D. Wark, J. Collette, J.Y. Reginster, Effects of long-term strontium ranelate treatment on vertebral fracture risk in postmenopausal women with osteoporosis, Osteoporos. Int. 20 (2009) 1663–1673. doi:10.1007/s00198-008-0825-6.

[56] P.J. Meunier, C. Roux, E. Seeman, S. Ortolani, J.E. Badurski, T.D. Spector, J. Cannata, A. Balogh, E.M. Lemmel, S. Pors-Nielsen, R. Rizzoli, H.K. Genant, J.Y. Reginster, The effects of strontium ranelate on the risk of vertebral fracture in women with postmenopausal osteoporosis, N. Engl. J. Med. 350 (2004) 459–468. doi:10.1056/NEJMoa022436.

[57] M.D. O’Donnell, P.L. Candarlioglu, C. a. Miller, E. Gentleman, M.M. Stevens, M.D. O’Donnell, P.L. Candarlioglu, C. a. Miller, E. Gentleman, M.M. Stevens, Materials characterisation and cytotoxic assessment of strontium-substituted bioactive glasses for bone regeneration, J. Mater. Chem. 20 (2010) 8934. doi:10.1039/c0jm01139h.

[58] E. Gentleman, Y.C. Fredholm, G. Jell, N. Lotfibakhshaiesh, M.D. O’Donnell, R.G. Hill, M.M. Stevens, The effects of strontium-substituted bioactive glasses on osteoblasts and osteoclasts in vitro, Biomaterials. 31 (2010) 3949–

Page 173: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 155

3956. doi:10.1016/j.biomaterials.2010.01.121. [59] S. Hesaraki, M. Alizadeh, H. Nazarian, D. Sharifi, Physico-chemical and in

vitro biological evaluation of strontium/calcium silicophosphate glass, J. Mater. Sci. Mater. Med. 21 (2010) 695–705. doi:10.1007/s10856-009-3920-0.

[60] M.E. Santocildes-Romero, A. Crawford, P. V Hatton, R.L. Goodchild, I.M. Reaney, C.A. Miller, The osteogenic response of mesenchymal stromal cells to strontium-substituted bioactive glasses, J. Tissue Eng. Regen. Med. 9 (2015) 619–631. doi:10.1002/term.2003.

[61] A.M. Barradas, H.A. Fernandes, N. Groen, Y.C. Chai, J. Schrooten, J. van de Peppel, J.P. van Leeuwen, C.A. van Blitterswijk, J. de Boer, A calcium-induced signaling cascade leading to osteogenic differentiation of human bone marrow-derived mesenchymal stromal cells, Biomaterials. 33 (2012) 3205–3215. doi:S0142-9612(12)00039-7 [pii]\r10.1016/j.biomaterials.2012.01.020.

[62] S. Peng, G. Zhou, K.D. Luk, K.M. Cheung, Z. Li, W.M. Lam, Z. Zhou, W.W. Lu, Strontium promotes osteogenic differentiation of mesenchymal stem cells through the Ras/MAPK signaling pathway, Cell Physiol Biochem. 23 (2009) 165–174. doi:000204105 [pii]\r10.1159/000204105.

[63] A. Di Martino, L. Liverani, A. Rainer, G. Salvatore, M. Trombetta, V. Denaro, Electrospun scaffolds for bone tissue engineering, Musculoskelet. Surg. 95 (2011) 69–80. doi:10.1007/s12306-011-0097-8.

[64] J.. Roether, a. . Boccaccini, L.. Hench, V. Maquet, S. Gautier, R. Jérôme, Development and in vitro characterisation of novel bioresorbable and bioactive composite materials based on polylactide foams and Bioglass® for tissue engineering applications, Biomaterials. 23 (2002) 3871–3878. doi:10.1016/S0142-9612(02)00131-X.

[65] D.M. Yunos, Z. Ahmad, V. Salih, a R. Boccaccini, Stratified scaffolds for osteochondral tissue engineering applications: electrospun PDLLA nanofibre coated Bioglass®-derived foams., J. Biomater. Appl. 27 (2013) 537–51. doi:10.1177/0885328211414941.

[66] S. Agarwal, J.H. Wendorff, A. Greiner, Use of electrospinning technique for biomedical applications, Polymer (Guildf). 49 (2008) 5603–5621. doi:10.1016/j.polymer.2008.09.014.

[67] J.J. Blaker, J.E. Gough, V. Maquet, I. Notingher, A.R. Boccaccini, In vitro evaluation of novel bioactive composites based on Bioglass®-filled polylactide foams for bone tissue engineering scaffolds, J. Biomed. Mater. Res. Part A. 67A (2003) 1401–1411. doi:10.1002/jbm.a.20055.

[68] H.H. Lu, S.F. El-Amin, K.D. Scott, C.T. Laurencin, Three-dimensional, bioactive, biodegradable, polymer–bioactive glass composite scaffolds with improved mechanical properties support collagen synthesis and mineralization of human osteoblast-like cells in vitro, J. Biomed. Mater. Res. Part A. 64A (2003) 465–474. doi:10.1002/jbm.a.10399.

[69] S.K. Misra, D. Mohn, T.J. Brunner, W.J. Stark, S.E. Philip, I. Roy, V. Salih, J.C. Knowles, A.R. Boccaccini, Comparison of nanoscale and microscale bioactive glass on the properties of P(3HB)/Bioglass® composites, Biomaterials. 29 (2008) 1750–1761. doi:10.1016/j.biomaterials.2007.12.040.

[70] A.R. Boccaccini, M. Erol, W.J. Stark, D. Mohn, Z. Hong, J.F. Mano,

Page 174: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

156 References

Polymer/bioactive glass nanocomposites for biomedical applications: A review, Compos. Sci. Technol. 70 (2010) 1764–1776. doi:10.1016/j.compscitech.2010.06.002.

[71] H.W. Kim, H.E. Kim, J.C. Knowles, Production and potential of bioactive glass nanofibers as a next-generation biomaterial, Adv. Funct. Mater. 16 (2006) 1529–1535.

[72] I. Armentano, M. Dottori, E. Fortunati, S. Mattioli, J.M. Kenny, Biodegradable polymer matrix nanocomposites for tissue engineering: A review, Polym. Degrad. Stab. 95 (2010) 2126–2146.

[73] H.W. Kim, H.H. Lee, G.S. Chun, Bioactivity and osteoblast responses of novel biomedical nanocomposites of bioactive glass nanofiber filled poly(lactic acid), J. Biomed. Mater. Res. A. 85 (2008) 651–663. doi:10.1002/jbm.a.31339.

[74] P. Fabbri, V. Cannillo, A. Sola, A. Dorigato, F. Chiellini, Highly porous polycaprolactone-45S5 Bioglass?? scaffolds for bone tissue engineering, Compos. Sci. Technol. 70 (2010) 1869–1878. doi:10.1016/j.compscitech.2010.05.029.

[75] J.-H. Jo, E.-J. Lee, D.-S. Shin, H.-E. Kim, H.-W. Kim, Y.-H. Koh, J.-H. Jang, In vitro/in vivo biocompatibility and mechanical properties of bioactive glass nanofiber and poly(ε-caprolactone) composite materials, J. Biomed. Mater. Res. Part B Appl. Biomater. 91B (2009) 213–220. doi:10.1002/jbm.b.31392.

[76] M.J. Rowe, K. Kamocki, D. Pankajakshan, D. Li, A. Bruzzaniti, V. Thomas, S.B. Blanchard, M.C. Bottino, Dimensionally stable and bioactive membrane for guided bone regeneration: An in vitro study, J. Biomed. Mater. Res. - Part B Appl. Biomater. 104 (2016) 594–605. doi:10.1002/jbm.b.33430.

[77] P.S.P. Poh, D.W. Hutmacher, B.M. Holzapfel, A.K. Solanki, M.M. Stevens, M.A. Woodruff, In vitro and in vivo bone formation potential of surface calcium phosphate-coated polycaprolactone and polycaprolactone/bioactive glass composite scaffolds, Acta Biomater. (2015). doi:10.1016/j.actbio.2015.11.012.

[78] J.M. Holzwarth, P.X. Ma, Biomimetic nanofibrous scaffolds for bone tissue engineering, Biomaterials. 32 (2011) 9622–9629. doi:10.1016/j.biomaterials.2011.09.009.

[79] A. Martins, J. V Araújo, R.L. Reis, N.M. Neves, Electrospun nanostructured scaffolds for tissue engineering applications., Nanomedicine (Lond). 2 (2007) 929–942. doi:10.2217/17435889.2.6.929.

[80] Q.P. Pham, U. Sharma, A.G. Mikos, Electrospinning of polymeric nanofibers for tissue engineering applications: a review, Tissue Eng. 12 (2006) 1197–1211. doi:10.1089/ten.2006.12.1197.

[81] M.C. Phipps, W.C. Clem, J.M. Grunda, G.A. Clines, S.L. Bellis, Increasing the pore sizes of bone-mimetic electrospun scaffolds comprised of polycaprolactone, collagen I and hydroxyapatite to enhance cell infiltration, Biomaterials. 33 (2012) 524–534. doi:10.1016/j.biomaterials.2011.09.080.

[82] R. Murugan, S. Ramakrishna, Nano-featured scaffolds for tissue engineering: a review of spinning methodologies, Tissue Eng. 12 (2006) 435–447. doi:10.1089/ten.2006.12.435.

Page 175: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 157

[83] D.W. Hutmacher, P.D. Dalton, Melt electrospinning, Chem. Asian J. 6 (2011) 44–56. doi:10.1002/asia.201000436.

[84] J. Stanger, N. Tucker, M. Staiger, Electrospinning, Smithers Rapra Technology, 2009. http://books.google.com.au/books?id=w_NjPgAACAAJ.

[85] J. Doshi, D.H. Reneker, Electrospinning process and applications of electrospun fibers, Conf. Rec. 1993 IEEE Ind. Appl. Conf. Twenty-Eighth IAS Annu. Meet. 35 (1993) 151–160. doi:10.1109/IAS.1993.299067.

[86] D.W. Hutmacher, P.D. Dalton, Melt electrospinning., Chem. Asian J. 6 (2011) 44–56. doi:10.1002/asia.201000436.

[87] R. Sahay, V. Thavasi, S. Ramakrishna, Design modifications in electrospinning setup for advanced applications, J. Nanomater. 2011 (2011). doi:10.1155/2011/317673.

[88] a. Cipitria, a. Skelton, T.R. Dargaville, P.D. Dalton, D.W. Hutmacher, Design, fabrication and characterization of PCL electrospun scaffolds—a review, J. Mater. Chem. 21 (2011) 9419. doi:10.1039/c0jm04502k.

[89] J. Nam, Y. Huang, S. Agarwal, J. Lannutti, Materials selection and residual solvent retention in biodegradable electrospun fibers, J. Appl. Polym. Sci. 107 (2008) 1547–1554. doi:10.1002/app.27063.

[90] T.D. Brown, P.D. Dalton, D.W. Hutmacher, Direct writing by way of melt electrospinning, Adv. Mater. 23 (2011) 5651–5657. doi:10.1002/adma.201103482.

[91] Y.M. Shin, M.M. Hohman, M.P. Brenner, G.C. Rutledge, Electrospinning: A whipping fluid jet generates submicron polymer fibers, Appl. Phys. Lett. 78 (2001) 1149–1151.

[92] A. Jaklenec, A. Stamp, E. Deweerd, A. Sherwin, R. Langer, Progress in the Tissue Engineering and Stem Cell Industry “Are we there yet?,” Tissue Eng. Part B. Rev. (2012). doi:10.1089/ten.TEB.2011.0553.

[93] T.D. Brown, F. Edin, N. Detta, A.D. Skelton, D.W. Hutmacher, P.D. Dalton, Melt electrospinning of poly(ε-caprolactone) scaffolds: Phenomenological observations associated with collection and direct writing, Mater. Sci. Eng. C. (2014). doi:10.1016/j.msec.2014.07.034.

[94] M.L. Muerza-Cascante, D. Haylock, D.W. Hutmacher, P.D. Dalton, Melt Electrospinning and Its Technologization in Tissue Engineering., Tissue Eng. Part B. Rev. 0 (2014) 1–16. doi:10.1089/ten.TEB.2014.0347.

[95] Y. Il Yoon, K.E. Park, S.J. Lee, W.H. Park, Fabrication of microfibrous and nano-/microfibrous scaffolds: Melt and hybrid electrospinning and surface modification of poly(L-lactic acid) with plasticizer, Biomed Res. Int. 2013 (2013). doi:10.1155/2013/309048.

[96] D.W. Hutmacher, P.D. Dalton, Melt electrospinning, Chem. - An Asian J. 6 (2011) 44–56. doi:10.1002/asia.201000436.

[97] S.R. Givens, K.H. Gardner, J.F. Rabolt, D.B. Chase, High-Temperature Electrospinning of Polyethylene Microfibers from Solution, Macromolecules. 40 (2007) 608–610. doi:10.1021/ma062398a.

[98] T. Yoshioka, R. Dersch, M. Tsuji, A.K. Schaper, Orientation analysis of individual electrospun PE nanofibers by transmission electron microscopy,

Page 176: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

158 References

Polymer (Guildf). 51 (2010) 2383–2389. doi:10.1016/j.polymer.2010.03.031. [99] H. Dinopoulos, R. Dimitriou, P. V Giannoudis, Bone graft substitutes: What

are the options?, Surgeon. 10 (2012) 230–9. doi:10.1016/j.surge.2012.04.001. [100] J. Henkel, M. Woodruff, D. Epari, R. Steck, V. Glatt, I. Dickinson, P. Choong,

M. Schuetz, D. Hutmacher, Bone Regeneration Based on Tissue Engineering Conceptions – A 21st Century Perspective, Bone Res. 1 (2013) 216–248. doi:10.4248/BR201303002.

[101] a. I. Pearce, R.G. Richards, S. Milz, E. Schneider, S.G. Pearce, Animal models for implant biomaterial research in bone: A review, Eur. Cells Mater. 13 (2007) 1–10. doi:vol013a01 [pii].

[102] A. Berner, J.D. Boerckel, S. Saifzadeh, R. Steck, J. Ren, C. Vaquette, J.Q. Zhang, M. Nerlich, R.E. Guldberg, D.W. Hutmacher, M.A. Woodruff, Biomimetic tubular nanofiber mesh and platelet rich plasma-mediated delivery of BMP-7 for large bone defect regeneration, Cell Tissue Res. 347 (2012) 603–12. doi:10.1007/s00441-011-1298-z.

[103] J.C. Reichert, A. Cipitria, D.R. Epari, S. Saifzadeh, P. Krishnakanth, A. Berner, M.A. Woodruff, H. Schell, M. Mehta, M.A. Schuetz, G.N. Duda, D.W. Hutmacher, A Tissue Engineering Solution for Segmental Defect Regeneration in Load-Bearing Long Bones, Sci. Transl. Med. 4 (2012) 141ra93-141ra93. doi:10.1126/scitranslmed.3003720.

[104] A. Berner, J.C. Reichert, M.A. Woodruff, S. Saifzadeh, A.J. Morris, D.R. Epari, M. Nerlich, M.A. Schuetz, D.W. Hutmacher, Autologous vs. allogenic mesenchymal progenitor cells for the reconstruction of critical sized segmental tibial bone defects in aged sheep, Acta Biomater. 9 (2013) 7874–84. doi:10.1016/j.actbio.2013.04.035.

[105] G.F. Muschler, V.P. Raut, T.E. Patterson, J.C. Wenke, J.O. Hollinger, The design and use of animal models for translational research in bone tissue engineering and regenerative medicine., Tissue Eng. Part B. Rev. 16 (2010) 123–145. doi:10.1089/ten.teb.2009.0658.

[106] M. a. Woodruff, C. Lange, J. Reichert, A. Berner, F. Chen, P. Fratzl, J.-T.T. Schantz, D.W. Hutmacher, Bone tissue engineering: from bench to bedside, Mater. Today. 15 (2012) 430–435. doi:10.1016/S1369-7021(12)70194-3.

[107] a. Berner, M. a. Woodruff, C.X.F. Lam, M.T. Arafat, S. Saifzadeh, R. Steck, J. Ren, M. Nerlich, a. K. Ekaputra, I. Gibson, D.W. Hutmacher, Effects of scaffold architecture on cranial bone healing., Int. J. Oral Maxillofac. Surg. 43 (2014) 506–13. doi:10.1016/j.ijom.2013.05.008.

[108] J.A. Ramos-Vara, M.A. Miller, When Tissue Antigens and Antibodies Get Along: Revisiting the Technical Aspects of Immunohistochemistry--The Red, Brown, and Blue Technique, Vet. Pathol. 51 (2014) 42–87. doi:10.1177/0300985813505879.

[109] J.D. Bancroft, M. Gamble, Theory and Practice of Histological Techniques, Churchill Livingstone, 2008. http://books.google.com.au/books?id=Dhn2KispfdQC.

[110] R. Yang, C.M. Davies, C.W. Archer, R.G. Richards, Immunohistochemistry of matrix markers in Technovit 9100 New-embedded undecalcified bone sections., Eur. Cell. Mater. 6 (2003) 57–71; discussion 71.

Page 177: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 159

http://www.ncbi.nlm.nih.gov/pubmed/14722903. [111] T. Goldschlager, A. Abdelkader, J. Kerr, I. Boundy, G. Jenkin, Undecalcified

bone preparation for histology, histomorphometry and fluorochrome analysis., J. Vis. Exp. (2010) 9–11. doi:10.3791/1707.

[112] G.R. Dickson, Methods of Calcified Tissue Preparation, Elsevier, 1984. [113] M. Villanueva-gomori, M. Villanueva-goldner, Part l. Decalcification, (1987). [114] N. Keklikoglu, S. Akinci, Comparison of three different techniques for

histological tooth preparation., Folia Histochem. Cytobiol. 51 (2013) 286–91. doi:10.5603/FHC.2013.0039.

[115] Y. Shibata, S. Fujita, H. Takahashi, a. Yamaguchi, T. Koji, Assessment of decalcifying protocols for detection of specific RNA by non-radioactive in situ hybridization in calcified tissues, Histochem. Cell Biol. 113 (2000) 153–159. doi:10.1007/s004180050434.

[116] M. Yoshioka, Y. Yoshida, S. Inoue, P. Lambrechts, G. Vanherle, Y. Nomura, M. Okazaki, H. Shintani, B. Van Meerbeek, B. Van Meerbeek, Adhesion/decalcification mechanisms of acid interactions with human hard tissues., J. Biomed. Mater. Res. 59 (2002) 56–62.

[117] P.J. Emans, S.K. Bulstra, R. Kuijer, The effects of different decalcification protocols on TUNEL and general cartilage staining., Biotech. Histochem. 80 (2009) 111–5. doi:10.1080/10520290500159253.

[118] D.L. Pitol, J.P.M. Issa, F.H. Caetano, L.O. Lunardi, Decalcification Dynamic of Dog Mineralized Tissue by Microwaves, Int. J. Morphol. 25 (2007) 309–313. doi:10.4067/S0717-95022007000200011.

[119] H.J. Helminen, Histochemistry, 127 (1980) 119–127. [120] D. Humphrey, F.E. Pittman, A simple methylene blue-azure 11-basic fuchsin

stain for epoxy-embedded tissue sections charles, 49 (1974) 9–14. [121] J. Bernhards, B. Weitzel, M. Werner, M. Rimpler, A. Georgii, A new

histological embedding method by low-temperature polymerisation of methyl methacrylate allowing immuno- and enzyme histochemical studies on semi-thin sections of undecalcified bone marrow biopsies, Histochemistry. 98 (1992) 145–154. doi:10.1007/BF00315873.

[122] R.G. Erben, Embedding of Bone Samples in Methylmethacrylate: An Improved Method Suitable for Bone Histomorphometry, Histochemistry, and Immunohistochemistry, J. Histochem. Cytochem. 45 (1997) 307–313. doi:10.1177/002215549704500215.

[123] G. Wittenburg, C. Volkel, R. Mai, G. Lauer, Immunohistochemical comparison of differentiation markers on paraffin and plastic embedded human bone samples., J. Physiol. Pharmacol. 60 Suppl 8 (2009) 43–9.

[124] C. Rentsch, W. Schneiders, S. Manthey, B. Rentsch, S. Rammelt, Comprehensive histological evaluation of bone implants., Biomatter. 4 (2014) 1–11. doi:10.4161/biom.27993.

[125] C. Rentsch, W. Schneiders, R. Hess, B. Rentsch, R. Bernhardt, K. Spekl, K. Schneider, D. Scharnweber, A. Biewener, S. Rammelt, Healing properties of surface-coated polycaprolactone-co-lactide scaffolds: a pilot study in sheep., J. Biomater. Appl. 28 (2014) 654–66. doi:10.1177/0885328212471409.

Page 178: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

160 References

[126] P.C. Pastoureau, E.B. Hunziker, J.-P. Pelletier, Cartilage, bone and synovial histomorphometry in animal models of osteoarthritis., Osteoarthritis Cartilage. 18 Suppl 3 (2010) S106-12. doi:10.1016/j.joca.2010.05.024.

[127] a Jäger, Histomorphometric study of age-related changes in remodelling activity of human desmodontal bone., J. Anat. 189 ( Pt 2 (1996) 257–64. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1167742&tool=pmcentrez&rendertype=abstract.

[128] C.M. Davies, D.B. Jones, M.J. Stoddart, K. Koller, E. Smith, C.W. Archer, R.G. Richards, Mechanically loaded ex vivo bone culture system “Zetos”: systems and culture preparation., Eur. Cell. Mater. 11 (2006) 57–75; discussion 75. http://www.ncbi.nlm.nih.gov/pubmed/16612792.

[129] M. Berger-Gorbet, B. Broxup, C. Rivard, L.H. Yahia, Biocompatibility testing of NiTi screws using immunohistochemistry on sections containing metallic implants., J. Biomed. Mater. Res. 32 (1996) 243–8. doi:10.1002/(SICI)1097-4636(199610)32:2&lt;243::AID-JBM14&gt;3.0.CO;2-K.

[130] U. Mayr-Wohlfart, G. Ravalli, Staining undecalcified bone sections a modified technique for an improved visualization of synthetic bone substitutes., Acta Chir. …. (2008) 443–445.

[131] Y. Förster, C. Rentsch, W. Schneiders, R. Bernhardt, J.C. Simon, H. Worch, S. Rammelt, Surface modification of implants in long bone., Biomatter. 2 (2012) 149–57. doi:10.4161/biom.21563.

[132] M.C. Schulz, A. Lode, S. Wittig, B. Stadlinger, E. Kuhlisch, U. Eckelt, M. Gelinsky, R. Mai, Characterization of the osseointegration of Algipore and Algipore modified with mineralized collagen type I., Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 114 (2012) S160-6. doi:10.1016/j.oooo.2011.09.029.

[133] T.D. Sargeant, M.O. Guler, S.M. Oppenheimer, A. Mata, R.L. Satcher, D.C. Dunand, S.I. Stupp, Hybrid bone implants: self-assembly of peptide amphiphile nanofibers within porous titanium., Biomaterials. 29 (2008) 161–71. doi:10.1016/j.biomaterials.2007.09.012.

[134] G. Hillmann, B. Hillman, K. Donath, Enzyme, Lectin and Immunohistochemistry of Plastic Embedded Undecalcified Bone and other Hard Tissues for Light Microscopic Investigations, 1982 (2000) 185–193.

[135] Y.-T. Kim, J.-K. Cha, J.-C. Park, U.-W. Jung, C.-S. Kim, K.-S. Cho, S.-H. Choi, In situ dental implant installation after decontamination in a previously peri-implant diseased site: a pilot study., J. Periodontal Implant Sci. 42 (2012) 13–9. doi:10.5051/jpis.2012.42.1.13.

[136] F. Butz, T. Ogawa, T.-L. Chang, I. Nishimura, Three-dimensional bone-implant integration profiling using micro-computed tomography., Int. J. Oral Maxillofac. Implants. 21 (2005) 687–695.

[137] H.E. Gruber, Adaptations of Goldner’s Masson Trichrome Stain for the Study of Undecalcified Plastic Embedded Bone, (2009).

[138] W. Schneiders, A. Reinstorf, M. Ruhnow, S. Rehberg, J. Heineck, I. Hinterseher, A. Biewener, H. Zwipp, S. Rammelt, Effect of chondroitin sulphate on material properties and bone remodelling around hydroxyapatite/collagen composites., J. Biomed. Mater. Res. A. 85 (2008) 638–45. doi:10.1002/jbm.a.31611.

Page 179: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 161

[139] U. Ripamonti, R.M. Klar, L.F. Renton, C. Ferretti, Synergistic induction of bone formation by hOP-1, hTGF-beta3 and inhibition by zoledronate in macroporous coral-derived hydroxyapatites., Biomaterials. 31 (2010) 6400–10. doi:10.1016/j.biomaterials.2010.04.037.

[140] S.-W. Kang, J.-S. Kim, K.-S. Park, B.-H. Cha, J.-H. Shim, J.Y. Kim, D.-W. Cho, J.-W. Rhie, S.-H. Lee, Surface modification with fibrin/hyaluronic acid hydrogel on solid-free form-based scaffolds followed by BMP-2 loading to enhance bone regeneration., Bone. 48 (2011) 298–306. doi:10.1016/j.bone.2010.09.029.

[141] Y.M. Shin, M.M. Hohman, M.P. Brenner, G.C. Rutledge, Electrospinning: A whipping fluid jet generates submicron polymer fibers, Appl. Phys. Lett. 78 (2001) 1149–1151. doi:10.1063/1.1345798.

[142] T.D. Brown, A. Slotosch, L. Thibaudeau, A. Taubenberger, D. Loessner, C. Vaquette, P.D. Dalton, D.W. Hutmacher, Design and fabrication of tubular scaffolds via direct writing in a melt electrospinning mode, Biointerphases. 7 (2012) 13. doi:10.1007/s13758-011-0013-7.

[143] R. Langer, J. Vacanti, Tissue engineering, Science (80-. ). 260 (1993) 920–926. doi:10.1126/science.8493529.

[144] L.J. White, V. Hutter, H. Tai, S.M. Howdle, K.M. Shakesheff, The effect of processing variables on morphological and mechanical properties of supercritical CO2 foamed scaffolds for tissue engineering, Acta Biomater. 8 (2012) 61–71. doi:http://dx.doi.org/10.1016/j.actbio.2011.07.032.

[145] Y. Nishimura, A. Takasu, Y. Inai, T. Hirabayashi, Melt spinning of poly(L-lactic acid) and its biodegradability, J. Appl. Polym. Sci. 97 (2005) 2118–2124. doi:10.1002/app.21915.

[146] G. Nitya, G.T. Nair, U. Mony, K.P. Chennazhi, S. V Nair, In vitro evaluation of electrospun PCL/nanoclay composite scaffold for bone tissue engineering., J. Mater. Sci. Mater. Med. 23 (2012) 1749–61. doi:10.1007/s10856-012-4647-x.

[147] X. Xiao, R. Liu, Q. Huang, X. Ding, Preparation and characterization of hydroxyapatite/polycaprolactone-chitosan composites., J. Mater. Sci. Mater. Med. 20 (2009) 2375–83. doi:10.1007/s10856-009-3810-5.

[148] L.L. Hench, R.J. Splinter, W.C. Allen, T.K. Greenlee, Bonding mechanisms at the interface of ceramic prosthetic materials, J. Biomed. Mater. Res. 5 (1971) 117–141. doi:10.1002/jbm.820050611.

[149] M. Brink, The influence of alkali and alkaline earths on the working range for bioactive glasses, J. Biomed. Mater. Res. 36 (1997) 109–117. doi:10.1002/(SICI)1097-4636(199707)36:1<109::AID-JBM13>3.0.CO;2-D.

[150] N.W. Marion, W. Liang, G.C. Reilly, D.E. Day, M.N. Rahaman, J.J. Mao, Borate Glass Supports the In Vitro Osteogenic Differentiation of Human Mesenchymal Stem Cells, Mech. Adv. Mater. Struct. 12 (2005) 239–246. doi:10.1080/15376490590928615.

[151] Y.C. Fredholm, N. Karpukhina, D.S. Brauer, J.R. Jones, R. V Law, R.G. Hill, Influence of strontium for calcium substitution in bioactive glasses on degradation, ion release and apatite formation., J. R. Soc. Interface. 9 (2012) 880–9. doi:10.1098/rsif.2011.0387.

Page 180: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

162 References

[152] A.S. Stephens, S.R. Stephens, C. Hobbs, D.W. Hutmacher, D. Bacic-Welsh, M.A. Woodruff, N.A. Morrison, Myocyte enhancer factor 2C, an osteoblast transcription factor identified by dimethyl sulfoxide (DMSO)-enhanced mineralization, J. Biol. Chem. 286 (2011) 30071–30086. doi:10.1074/jbc.M111.253518.

[153] K.H. Jones, J. a Senft, An improved method to determine cell viability by simultaneous staining with fluorescein diacetate-propidium iodide., J. Histochem. Cytochem. 33 (1985) 77–79. doi:10.1177/33.1.2578146.

[154] K. a Blackwood, R. McKean, I. Canton, C.O. Freeman, K.L. Franklin, D. Cole, I. Brook, P. Farthing, S. Rimmer, J.W. Haycock, A.J. Ryan, S. MacNeil, Development of biodegradable electrospun scaffolds for dermal replacement., Biomaterials. 29 (2008) 3091–104. doi:10.1016/j.biomaterials.2008.03.037.

[155] S. Labbaf, O. Tsigkou, K.H. Müller, M.M. Stevens, A.E. Porter, J.R. Jones, Spherical bioactive glass particles and their interaction with human mesenchymal stem cells in vitro., Biomaterials. 32 (2011) 1010–8. doi:10.1016/j.biomaterials.2010.08.082.

[156] C. a Gregory, W.G. Gunn, A. Peister, D.J. Prockop, An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction., Anal. Biochem. 329 (2004) 77–84. doi:10.1016/j.ab.2004.02.002.

[157] V.M.C. Quent, D. Loessner, T. Friis, J.C. Reichert, D.W. Hutmacher, Discrepancies between metabolic activity and DNA content as tool to assess cell proliferation in cancer research., J. Cell. Mol. Med. 14 (2010) 1003–13. doi:10.1111/j.1582-4934.2010.01013.x.

[158] L.D. Shea, D. Wang, R.T. Franceschi, D.J. Mooney, Engineered bone development from a pre-osteoblast cell line on three-dimensional scaffolds., Tissue Eng. 6 (2000) 605–617. doi:10.1089/10763270050199550.

[159] P. Han, C. Wu, J. Chang, Y. Xiao, The cementogenic differentiation of periodontal ligament cells via the activation of Wnt/β-catenin signalling pathway by Li+ ions released from bioactive scaffolds., Biomaterials. 33 (2012) 6370–9. doi:10.1016/j.biomaterials.2012.05.061.

[160] S. Fu, P. Ni, B. Wang, B. Chu, J. Peng, L. Zheng, X. Zhao, F. Luo, Y. Wei, Z. Qian, In vivo biocompatibility and osteogenesis of electrospun poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone)/nano-hydroxyapatite composite scaffold., Biomaterials. 33 (2012) 8363–71. doi:10.1016/j.biomaterials.2012.08.023.

[161] F. Chen, Z.C. Wang, C.J. Lin, Preparation and characterization of nano-sized hydroxyapatite particles and hydroxyapatite/chitosan nano-composite for use in biomedical materials, Mater. Lett. 57 (2002) 858–861. doi:10.1016/S0167-577X(02)00885-6.

[162] H. Kim, H. Lee, J.C. Knowles, Electrospinning biomedical nanocomposite fibers of hydroxyapaite / poly ( lactic acid ) for bone regeneration, (2006). doi:10.1002/jbm.a.

[163] H.-W. Kim, H.-H. Lee, J.C. Knowles, Electrospinning biomedical nanocomposite fibers of hydroxyapatite/poly(lactic acid) for bone regeneration, J. Biomed. Mater. Res. Part A. 79A (2006) 643–649.

Page 181: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 163

doi:10.1002/jbm.a.30866. [164] Q.P. Pham, U. Sharma, A.G. Mikos, Electrospun Poly(ε-caprolactone)

Microfiber and Multilayer Nanofiber/Microfiber Scaffolds:  Characterization of Scaffolds and Measurement of Cellular Infiltration, Biomacromolecules. 7 (2006) 2796–2805. doi:10.1021/bm060680j.

[165] M. Ebrahimian-Hosseinabadi, F. Ashrafizadeh, M. Etemadifar, S.S. Venkatraman, Preparation and mechanical behavior of PLGA/nano-BCP composite scaffolds during in-vitro degradation for bone tissue engineering, Polym. Degrad. Stab. 96 (2011) 1940–1946. doi:10.1016/j.polymdegradstab.2011.05.016.

[166] J.-H. Jo, E.-J. Lee, D.-S. Shin, H.-E. Kim, H.-W. Kim, Y.-H. Koh, J.-H. Jang, In vitro/in vivo biocompatibility and mechanical properties of bioactive glass nanofiber and poly(epsilon-caprolactone) composite materials., J. Biomed. Mater. Res. B. Appl. Biomater. 91 (2009) 213–20. doi:10.1002/jbm.b.31392.

[167] M. Persson, G.S. Lorite, S.-W. Cho, J. Tuukkanen, M. Skrifvars, Melt spinning of poly(lactic acid) and hydroxyapatite composite fibers: influence of the filler content on the fiber properties., ACS Appl. Mater. Interfaces. 5 (2013) 6864–72. doi:10.1021/am401895f.

[168] P.J. Marie, P. Ammann, G. Boivin, C. Rey, Mechanisms of action and therapeutic potential of strontium in bone, Calcif. Tissue Int. 69 (2001) 121–129. doi:10.1007/s002230010055.

[169] S.C. Verberckmoes, M.E. De Broe, P.C. D’Haese, Dose-dependent effects of strontium on osteoblast function and mineralization, Kidney Int. 64 (2003) 534–543. doi:10.1046/j.1523-1755.2003.00123.x.

[170] K. Qiu, X.J. Zhao, C.X. Wan, C.S. Zhao, Y.W. Chen, Effect of strontium ions on the growth of ROS17/2.8 cells on porous calcium polyphosphate scaffolds, Biomaterials. 27 (2006) 1277–1286. doi:10.1016/j.biomaterials.2005.08.006.

[171] C. Wu, Y. Zhou, C. Lin, J. Chang, Y. Xiao, Strontium-containing mesoporous bioactive glass scaffolds with improved osteogenic/cementogenic differentiation of periodontal ligament cells for periodontal tissue engineering., Acta Biomater. 8 (2012) 3805–15. doi:10.1016/j.actbio.2012.06.023.

[172] C. Wu, W. Fan, M. Gelinsky, Y. Xiao, P. Simon, R. Schulze, T. Doert, Y. Luo, G. Cuniberti, Bioactive SrO-SiO2 glass with well-ordered mesopores: Characterization, physiochemistry and biological properties, Acta Biomater. 7 (2011) 1797–1806. doi:10.1016/j.actbio.2010.12.018.

[173] J. Liu, S.C.F. Rawlinson, R.G. Hill, F. Fortune, Strontium-substituted bioactive glasses in vitro osteogenic and antibacterial effects, Dent. Mater. 32 (2016) 412–422. doi:10.1016/j.dental.2015.12.013.

[174] C. Wu, Y. Zhou, W. Fan, P. Han, J. Chang, J. Yuen, M. Zhang, Y. Xiao, Hypoxia-mimicking mesoporous bioactive glass scaffolds with controllable cobalt ion release for bone tissue engineering, Biomaterials. 33 (2012) 2076–2085. doi:10.1016/j.biomaterials.2011.11.042.

[175] S.-H. Shin, O. Purevdorj, O. Castano, J. a Planell, H.-W. Kim, A short review: Recent advances in electrospinning for bone tissue regeneration., J. Tissue Eng. 3 (2012) 2041731412443530. doi:10.1177/2041731412443530.

Page 182: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

164 References

[176] A. Takeuchi, C. Ohtsuki, T. Miyazaki, H. Tanaka, M. Yamazaki, M. Tanihara, Deposition of bone-like apatite on silk fiber in a solution that mimics extracellular fluid., J. Biomed. Mater. Res. A. 65 (2003) 283–289. doi:10.1002/jbm.a.10456.

[177] W.L. Murphy, D.H. Kohn, D.J. Mooney, Growth of continuous bonelike mineral within porous poly(lactide-co-glycolide) scaffolds in vitro., J. Biomed. Mater. Res. 50 (2000) 50–8. doi:10.1002/(SICI)1097-4636(200004)50:1<50::AID-JBM8>3.0.CO;2-F.

[178] F. Yang, J.G.C. Wolke, J. a. Jansen, Biomimetic calcium phosphate coating on electrospun poly(ɛ-caprolactone) scaffolds for bone tissue engineering, Chem. Eng. J. 137 (2008) 154–161. doi:10.1016/j.cej.2007.07.076.

[179] X. Li, J. Xie, X. Yuan, Y. Xia, Coating electrospun poly(epsilon-caprolactone) fibers with gelatin and calcium phosphate and their use as biomimetic scaffolds for bone tissue engineering., Langmuir. 24 (2008) 14145–50. doi:10.1021/la802984a.

[180] M. Yong, F. Melchels, C. Vaquette, D. Hutmacher, C. Adam, M. Domingos, P. Bartolo, Polycaprolactone-based scaffold plus BMP-2 in a sheep thoracic spine fusion model, in: Innov. Dev. Virtual Phys. Prototyp. - Proc. 5th Int. Conf. Adv. Res. Rapid Prototyp., 2012: pp. 89–92.

[181] H. Jones, An Improved Method Simultaneous Staining to Determine Cell with Fluorescein Iodide ’ Viability by, (1985).

[182] D. Cao, Y.P. Wu, Z.F. Fu, Y. Tian, C.J. Li, C.Y. Gao, Z.L. Chen, X.Z. Feng, Cell adhesive and growth behavior on electrospun nanofibrous scaffolds by designed multifunctional composites, Colloids Surfaces B Biointerfaces. 84 (2011) 26–34. doi:10.1016/j.colsurfb.2010.12.005.

[183] P. Szulc, P. Garnero, F. Marchand, F. Duboeuf, P.D. Delmas, Biochemical markers of bone formation reflect endosteal bone loss in elderly men--MINOS study, Bone. 36 (2005) 13–21. doi:10.1016/j.bone.2004.09.004.

[184] R. J., L. I., S. S., L. A., N. K., H. A., H. T., M. R., K. C., S. M., Human mastoid periosteum-derived stem cells: Promising candidates for skeletal tissue engineering, J. Tissue Eng. Regen. Med. 2 (2008) 136–146. doi:10.1002/term.75.

[185] M. Taira, H. Nakao, J. Takahashi, Y. Araki, Effects of two vitamins, two growth factors and dexamethasone on the proliferation of rat bone marrow stromal cells and osteoblastic MC3T3-E1 cells, J. Oral Rehabil. 30 (2003) 697–701. doi:10.1046/j.1365-2842.2003.01118.x.

[186] F. Tan, M. Naciri, M. Al-Rubeai, Osteoconductivity and growth factor production by MG63 osteoblastic cells on bioglass-coated orthopedic implants., Biotechnol. Bioeng. 108 (2011) 454–64. doi:10.1002/bit.22955.

[187] V. Maquet, A.R. Boccaccini, L. Pravata, I. Notingher, R. Jérôme, Preparation, characterization, and in vitro degradation of bioresorbable and bioactive composites based on Bioglass-filled polylactide foams., J. Biomed. Mater. Res. A. 66 (2003) 335–346. doi:10.1002/jbm.a.10587.

[188] A.J. Aho, T. Tirri, J. Kukkonen, N. Strandberg, J. Rich, J. Seppala, A. Yli-Urpo, Injectable bioactive glass/biodegradable polymer composite for bone and cartilage reconstruction: concept and experimental outcome with

Page 183: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 165

thermoplastic composites of poly(epsilon-caprolactone-co-D,L-lactide) and bioactive glass S53P4., J. Mater. Sci. Mater. Med. 15 (2004) 1165–1173. doi:10.1023/B:JMSM.0000046401.50406.9b.

[189] B. Chen, K. Sun, Poly (??-caprolactone)/hydroxyapatite composites: Effects of particle size, molecular weight distribution and irradiation on interfacial interaction and properties, Polym. Test. 24 (2005) 64–70. doi:10.1016/j.polymertesting.2004.07.010.

[190] J. Lyons, C. Li, F. Ko, Melt-electrospinning part I: Processing parameters and geometric properties, Polymer (Guildf). 45 (2004) 7597–7603. doi:10.1016/j.polymer.2004.08.071.

[191] J. Ren, K. a. Blackwood, A. Doustgani, P.P. Poh, R. Steck, M.M. Stevens, M. a. Woodruff, Melt-electrospun polycaprolactone strontium-substituted bioactive glass scaffolds for bone regeneration, J. Biomed. Mater. Res. - Part A. 102 (2014) 3140–3153. doi:10.1002/jbm.a.34985.

[192] Q. Hou, D.W. Grijpma, J. Feijen, Porous polymeric structures for tissue engineering prepared by a coagulation, compression moulding and salt leaching technique, Biomaterials. 24 (2003) 1937–1947. doi:10.1016/S0142-9612(02)00562-8.

[193] N. Ristovski, N. Bock, S. Liao, S.K. Powell, J. Ren, G.T.S. Kirby, K.A. Blackwood, M.A. Woodruff, Improved fabrication of melt electrospun tissue engineering scaffolds using direct writing and advanced electric field control., Biointerphases. 10 (2015) 11006. doi:10.1116/1.4914380.

[194] H. Davis, J. Leach, Hybrid and Composite Biomaterials in Tissue Engineering, Top. Multifunct. Biomater. Devices. (2008) 1–26. http://www.oulu.fi/spareparts/ebook_topics_multifunctional/abstracts/davis.pdf.

[195] K.-J. Wu, C.-S. Wu, J.-S. Chang, Biodegradability and mechanical properties of polycaprolactone composites encapsulating phosphate-solubilizing bacterium Bacillus sp. PG01, Process Biochem. 42 (2007) 669–675. doi:10.1016/j.procbio.2006.12.009.

[196] C.-S. Wu, Physical properties and biodegradability of maleated-polycaprolactone/starch composite, Polym. Degrad. Stab. 80 (2003) 127–134. doi:10.1016/S0141-3910(02)00393-2.

[197] E. Di Maio, S. Iannace, L. Sorrentino, L. Nicolais, Isothermal crystallization in PCL/clay nanocomposites investigated with thermal and rheometric methods, Polymer (Guildf). 45 (2004) 8893–8900. doi:10.1016/j.polymer.2004.10.037.

[198] P. Pokorny, E. Kostakova, F. Sanetrnik, P. Mikes, J. Chvojka, T. Kalous, M. Bilek, K. Pejchar, J. Valtera, D. Lukas, Effective AC needleless and collectorless electrospinning for yarn production, Phys. Chem. Chem. Phys. 16 (2014) 26816–26822. doi:10.1039/C4CP04346D.

[199] P. Roschger, P. Fratzl, J. Eschberger, K. Klaushofer, Validation of quantitative backscattered electron imaging for the measurement of mineral density distribution in human bone biopsies, Bone. 23 (1998) 319–326. doi:10.1016/S8756-3282(98)00112-4.

[200] R.D. Bloebaum, J.G. Skedros, E.G. Vajda, K.N. Bachus, B.R. Constantz,

Page 184: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

166 References

Determining mineral content variations in bone using backscattered electron imaging, Bone. 20 (1997) 485–490. doi:10.1016/S8756-3282(97)00015-X.

[201] B. Bracci, P. Torricelli, S. Panzavolta, E. Boanini, R. Giardino, A. Bigi, Effect of Mg2+, Sr2+, and Mn2+ on the chemico-physical and in vitro biological properties of calcium phosphate biomimetic coatings, J. Inorg. Biochem. 103 (2009) 1666–1674. doi:10.1016/j.jinorgbio.2009.09.009.

[202] G.S. Stein, J.B. Lian, A.J. van Wijnen, J.L. Stein, M. Montecino, A. Javed, S.K. Zaidi, D.W. Young, J.-Y. Choi, S.M. Pockwinse, Runx2 control of organization, assembly and activity of the regulatory machinery for skeletal gene expression., Oncogene. 23 (2004) 4315–4329. doi:10.1038/sj.onc.1207676.

[203] G.R. Beck, E.C. Sullivan, E. Moran, B. Zerler, Relationship between alkaline phosphatase levels, osteopontin expression, and mineralization in differentiating MC3T3-E1 osteoblasts, J. Cell. Biochem. 68 (1998) 269–280. doi:10.1002/(SICI)1097-4644(19980201)68:2<269::AID-JCB13>3.0.CO;2-A.

[204] G. V Guibas, S. Lakis, C. Gkimpas, M. Manda, D. Kapoukranidou, E. Spandou, Efficiency of different decalcification protocols for nasal osseous structures in a rat experimental model of allergic rhinitis, and their effects on epithelial histology: an attempt at standardization., Exp. Toxicol. Pathol. 66 (2014) 469–75. doi:10.1016/j.etp.2014.09.001.

[205] N.B. Bauer, T. El Khassawna, F. Goldmann, M. Stirn, D. Ledieu, G. Schlewitz, P. Govindarajan, D. Zahner, D. Weisweiler, N. Schliefke, W. Böcker, R. Schnettler, C. Heiss, A. Moritz, Characterization of bone turnover and energy metabolism in a rat model of primary and secondary osteoporosis., Exp. Toxicol. Pathol. 67 (2015) 287–96. doi:10.1016/j.etp.2015.01.004.

[206] S. Rammelt, C. Heck, R. Bernhardt, S. Bierbaum, D. Scharnweber, A. Biewener, H. Zwipp, In Vivo Effects of Coating Loaded and Unloaded Ti Implants with Collagen , Chondroitin Sulfate , and Hydroxyapatite in the Sheep Tibia, (2007) 1052–1061. doi:10.1002/jor.

[207] J.C. Reichert, A. Cipitria, D.R. Epari, S. Saifzadeh, P. Krishnakanth, A. Berner, M.A. Woodruff, H. Schell, M. Mehta, M.A. Schuetz, G.N. Duda, D.W. Hutmacher, P. Krishnakanth, A. Berner, M.A. Woodruff, H. Schell, M. Mehta, M.A. Schuetz, G.N. Duda, D.W. Hutmacher, A Tissue Engineering Solution for Segmental Defect Regeneration in Load-Bearing Long Bones, Sci. Transl. Med. 4 (2012) 141ra93. doi:10.1126/scitranslmed.3003720.

[208] R.P. Meinig, C.M. Buesing, J. Helm, S. Gogolewski, Regeneration of diaphyseal bone defects using resorbable poly(L/DL-lactide) and poly(D-lactide) membranes in the Yucatan pig model, J Orthop Trauma. 11 (1997) 551–558.

[209] P. De Coppi, G. Bartsch, M.M. Siddiqui, T. Xu, C.C. Santos, L. Perin, G. Mostoslavsky, A.C. Serre, E.Y. Snyder, J.J. Yoo, M.E. Furth, S. Soker, A. Atala, Isolation of amniotic stem cell lines with potential for therapy., Nat. Biotechnol. 25 (2007) 100–6. doi:10.1038/nbt1274.

[210] J. Cano-Sanchez, J. Campo-Trapero, J.C. Gonzalo-Lafuente, L.A. Moreno-Lopez, A. Bascones-Martinez, Undecalcified bone samples: a description of the technique and its utility based on the literature., Med. Oral Patol. Oral Cir. Bucal. 10 Suppl 1 (2005) E74-87.

Page 185: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

References 167

[211] M. McGee-Lawrence, D. Razidlo, Induction of Fully Stabilized Cortical Bone Defects to Study Intramembranous Bone Regeneration, in: J.J. Westendorf, A.J. van Wijnen (Eds.), Osteoporos. Osteoarthr. SE - 14, Springer New York, 2015: pp. 183–192. doi:10.1007/978-1-4939-1619-1_14.

[212] U.T. Iwaniec, T.J. Wronski, R.T. Turner, Histological Analysis of Bone , Alcohol . 447 (2008) 325–341. http://www.springerprotocols.com/Abstract/doi/10.1007/978-1-59745-242-7_21.

[213] T.B. Bemenderfer, J.S. Harris, K.W. Condon, M.A. Kacena, Tips and Techniques for Processing and Sectioning Undecalcified Murine Bone Specimens , Skelet. Dev. Repair . 1130 (2014) 123–147. http://www.springerprotocols.com/Abstract/doi/10.1007/978-1-62703-989-5_10.

[214] S. Rammelt, D. Corbeil, S. Manthey, H. Zwipp, U. Hanisch, Immunohistochemical in situ characterization of orthopedic implants on polymethyl metacrylate embedded cutting and grinding sections, (2007). doi:10.1002/jbm.a.

[215] E. Cruber, Adaptations of Goldner ’ s Masson Trichrome Stain for the Study of Undecalcified Plastic Embedded Bone, (1988) 30–34.

[216] G. Avwioro, HISTOCHEMICAL USES OF HAEMATOXYLIN - A REVIEW, 1 (2011) 24–34.

[217] V. Espina, J.D. Wulfkuhle, V.S. Calvert, A. VanMeter, W. Zhou, G. Coukos, D.H. Geho, E.F. Petricoin, L.A. Liotta, Laser-capture microdissection., Nat. Protoc. 1 (2006) 586–603. doi:10.1038/nprot.2006.85.

[218] K. Bohmann, G. Hennig, U. Rogel, C. Poremba, B.M. Mueller, P. Fritz, S. Stoerkel, K.L. Schaefer, RNA extraction from archival formalin-fixed paraffin-embedded tissue: A comparison of manual, semiautomated, and fully automated purification methods, Clin. Chem. 55 (2009) 1719–1727. doi:10.1373/clinchem.2008.122572.

[219] L.C. Lawrie, S. Curran, H.L. McLeod, J.E. Fothergill, G.I. Murray, Application of laser capture microdissection and proteomics in colon cancer., Mol. Pathol. 54 (2001) 253–258. doi:10.1136/mp.54.4.253.

[220] B.J. Xu, Combining laser capture microdissection and proteomics: Methodologies and clinical applications, Proteomics - Clin. Appl. 4 (2010) 116–123. doi:10.1002/prca.200900138.

[221] F. Von Eggeling, H. Davies, L. Lomas, W. Fiedler, K. Junker, U. Claussen, G. Ernst, Tissue-specific microdissection coupled with ProteinChip® array technologies: Applications in cancer research, Biotechniques. 29 (2000) 1066–1070.

[222] D.P.D. Burnett, J. Crocker, The science of laboratory diagnosis, 2nd ed., Wiley, Chichester, 2005.

[223] C.M. Langton, C.F. Njehs, J. Cameron, The Physical Measurement of Bone, Med. Phys. 31 (2004) 1648. doi:10.1118/1.1752929.

[224] G. Kirby, L. White, R. Steck, A. Berner, K. Bogoevski, O. Qutachi, B. Jones, S. Saifzadeh, D. Hutmacher, K. Shakesheff, M. Woodruff, Microparticles for Sustained Growth Factor Delivery in the Regeneration of Critically-Sized

Page 186: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

168 References

Segmental Tibial Bone Defects, Materials (Basel). 9 (2016) 259. doi:10.3390/ma9040259.

[225] A.G. Mikos, S.W. Herring, P. Ochareon, J. Elisseeff, H.H. Lu, R. Kandel, F.J. Schoen, M. Toner, D. Mooney, A. Atala, M.E. Van Dyke, D. Kaplan, G. Vunjak-Novakovic, Engineering complex tissues., Tissue Eng. 12 (2006) 3307–3339. doi:10.1089/ten.2006.12.3307.

[226] P.P. Spicer, J.D. Kretlow, S. Young, J.A. Jansen, F.K. Kasper, A.G. Mikos, Evaluation of bone regeneration using the rat critical size calvarial defect, Nat. Protoc. 7 (2012) 1918–1929. doi:10.1038/nprot.2012.113.

Page 187: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Appendices 169

Appendices

Appendix A

Figure A.1 Goldner’s trichrome staining of resin embedded sheep tibia bone tissues around titanium implant, sections obtained by ground sectioning technique [206]. The mineralized bone which should be green based on the staining protocol shows ‘patchy’ stains of green and red, thus weekending the differentiation between hard and soft tissues.

Page 188: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

170 Appendices

Figure A.2 Goldner’s trichrome staining of resin embedded sheep tibia native bone tissues and regenerated bone tissues around implanted PCL scaffolds , sections obtained by ground sectioning technique [125]. The images c and d showed inconsistent bone staining mixing green and red colours.

Page 189: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

Appendices 171

Figure A.3 Goldner’s trichrome staining of resin embedded sheep tibia bone tissues around Ti implant, sections obtained by ground sectioning technique [131]. Comparing image C and D, the fibrous tissue and bone tissue both stained red, which presents poor contrast between these two tissue types.

Page 190: DEVELOPMENT OF MELT ELECTROSPUN COMPOSITE … Edward_Ren_Thesis.pdf · bone defect regeneration, Cell and Tissue Research, 2012 2. A Berner, J D Boerckel, S Saifzadeh, R Steck, J

172 Appendices

Appendix B

SrBG particle grinding and sizing experiment

The bioactive glass particles were ground for 30 seconds, 1 minutes, 2 minutes, 4 minutes, 6 minutes, 8 minutes, 10 minutes, 12 minutes, 16 minutes, 20 minutes, 24 minutes, 30 minutes, 36 minutes, 42 minutes, 48 minutes, 54 minutes, 60 minutes, 66 minutes, 72 minutes, 84 minutes, 96 minutes and 108 minutes, 2 hours, 4 hours, 6 hours and 8 hours. Their size distribution was measured after each time point. The example results are shown in the figure below.

Volu

me

dens

ity (%

)

Particle size (μm)