Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering...

160
Modular Approach to Adipose Tissue Engineering by Mark James Butler A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Chemical Engineering and Applied Chemistry & Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Mark James Butler 2011

Transcript of Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering...

Page 1: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

Modular Approach to Adipose Tissue Engineering

by

Mark James Butler

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Graduate Department of Chemical Engineering and Applied Chemistry &

Institute of Biomaterials and Biomedical Engineering

University of Toronto

© Copyright by Mark James Butler 2011

Page 2: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

ii

Modular Approach to Adipose Tissue Engineering

Mark James Butler Doctor of Philosophy

Department of Chemical Engineering and Applied Chemistry University of Toronto, 2011

Abstract Despite the increasing clinical demand in reconstructive, cosmetic and correctional surgery there

remains no optimal strategy for the regeneration or replacement of adipose tissue. Previous

approaches to adipose tissue engineering have failed to create an adipose tissue depot that

maintains implant volume in vivo long-term (>3 months). This is due to inadequate mechanical

properties of the biomaterial and insufficient vascularization upon implantation. Modular tissue

engineering is a means to produce large volume functional tissues from small sub-mm sized

tissues with an intrinsic vascularization. We first explored the potential of a semi-synthetic

collagen/poloxamine hydrogel with improved mechanical properties to be used as the module

biomaterial. We found this biomaterial to not be suitable for adipose tissue engineering because

it did not support embedded adipose-derived stem cell (ASC) viability, differentiation and

human microvascular endothelial cell (HMEC) attachment. ASC-embedded collagen gel

modules coated with HMEC were then implanted subcutaneously in SCID mice to study its

revascularization potential. ASC cotransplantation was shown to drive HMEC vascularization in

vivo: HMEC were seen to detach from the surface of the modules to form vessels containing

erythrocytes as early as day 3; vessels decreased in number but increased in size over 14 days;

and persisted for up to 3 months. Early vascularization promoted fat development. Only in the

case of ASC-HMEC cotransplantation was progressive fat accumulation observed in the module

implants. Although implant volume was not maintained, likely due rapid collagen degradation,

the key result here is that ASC-HMEC cotransplantation in the modular approach was successful

Page 3: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

iii

in creating vascularized adipose tissue in vivo that persisted for 3 months. The modular system

was then studied in vitro to further understand ASC-EC interaction. Coculture with ASC was

shown to promote an angiogenic phenotype (e.g. sprouting, migration) from HUVEC on

modules. RT-PCR analysis revealed that VEGF, PAI-1 and TNFα was involved in ASC-EC

paracrine signalling. In summary, ASC-HMEC cotransplantation in modules was effective in

rapidly forming a vascular network that supported fat development. Future work should focus

on further elucidating ASC-EC interactions and developing a suitable biomaterial to improve

adipose tissue development and volume maintenance of engineered constructs.

Page 4: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

iv

Acknowledgements 

I first would like to thank Dr. Michael Sefton for his help and guidance through the years.

I have been a student in his lab from the beginning of my research experiences in my 4th year

undergraduate project. I remember waiting for our very first meeting outside his office trying to

digest my very first journal publication, thinking it was written in a language I did not

understand. I have come a long way over the years learning new terms, concepts, techniques,

and how to develop and test hypotheses. From Dr. Sefton I have learned two things: First,

effective research must tell a story and second, learning is a life long adventure that does not end

with the awarding of a PhD.

I thank all members of the Sefton lab, past and present, that contributed to my life both

academically and personally over the years. Thanks to Omar Khan, Brendan Leung, Lindsay

Fitzpatrick, Rohini Gupta, Dean Chamberlain, Ema Ciucurel, and Derek Voice for the many

brainstorming sessions, help with experimental work, research discussions and the times outside

of the lab that made graduate school so enjoyable. A special thanks to Chuen Lo for his

expertise and help with animal work.

I would like to thank all other collaborators on this project: Kim Woodhouse for her help

and guidance as a cosupervisor; Yu-Ling Cheng for being part of the reading committee; and

Alison McGuigan for her help early on in my research career as a 4th year thesis supervisor and

her mentorship over the years. A special thanks to Dr. John Semple and his colleague Dr. Bell

for the adipose tissue samples. Without your help this work would not have been possible.

Finally, I thank my family, my parents, Jim and Barb, and sisters, Sarah and Glenda for

being there for me and encouraging me to do my best every step of the way. Thank you to

Nicole and Kostas for listening to some of my crazy ideas and providing the needed distraction

from work. If you’ve made it this far, thanks to you for reading my thesis. It was a long journey

with some ups and downs, but such a worthwhile experience.

Page 5: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

v

Table of Contents 

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

Acknowledgements..............................................................................................................................iv

Table of Contents ................................................................................................................................... v

List of Figures.........................................................................................................................................ix

List of Tables ........................................................................................................................................ xii

Abbreviations...................................................................................................................................... xiii

1 Introduction, Hypothesis and Experimental Plan.............................................................1

1.1 Clinical Impetus .............................................................................................................. 1

1.2 Modular Tissue Engineering........................................................................................... 2

1.3 Objectives and Hypothesis.............................................................................................. 4

1.4 Thesis Organization ........................................................................................................ 5

2 Adipose Tissue Engineering & Adipose‐derived Stem Cell/Endothelial Cell 

Cotransplantation as a Revascularization Strategy...................................................................6

2.1 Adipose-derived Stem Cells and Soft Tissue Engineering............................................. 6

2.1.1 Adipose Tissue........................................................................................................ 6

2.1.2 Adipocyte Differentiation ....................................................................................... 8

2.1.3 Adipose Tissue Extracellular Matrix .................................................................... 12

2.1.4 Stem Cells for Adipose Tissue Engineering ......................................................... 13

2.1.5 Adipose Tissue Engineering Strategies ................................................................ 14

2.1.6 Semi-Synthetic Collagen/Poloxamine Hydrogel .................................................. 16

2.2 Angiogenesis & Revascularization Strategies .............................................................. 18

2.2.1 Mechanisms of Angiogenesis ............................................................................... 18

2.2.1.1 Vascular Functions in Adipose Tissues ............................................................ 21

2.2.1.2 Angiogenic Modulators .................................................................................... 22

2.2.1.3 Relationship between ASC and EC .................................................................. 24

2.2.2 Revascularization Strategies ................................................................................. 26

2.2.2.1 Growth Factor Delivery .................................................................................... 27

2.2.2.2 Biomaterial-mediated Angiogenesis................................................................. 28

Page 6: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

vi

2.2.2.3 Vascular Pedicle................................................................................................ 29

2.2.2.4 Cell-Based Revascularization Strategies .......................................................... 29

3 Semi‐Synthetic Collagen/Poloxamine Hydrogel as a Potential Biomaterial for 

Adipose Tissue Engineering............................................................................................................ 33

3.1 Abstract ......................................................................................................................... 33

3.2 Introduction................................................................................................................... 33

3.3 Materials and Methods.................................................................................................. 36

3.3.1 Cells ...................................................................................................................... 36

3.3.2 Hydrogel Synthesis ............................................................................................... 37

3.3.3 Cell Viability......................................................................................................... 38

3.3.4 Hydrogel Characterization .................................................................................... 39

3.3.5 Oil Red O .............................................................................................................. 39

3.3.6 Glycerol-3-phosphate Dehydrogenase Activity.................................................... 40

3.3.7 EC Attachment to Gels and Modules.................................................................... 40

3.3.8 Implantation of Collagen/Poloxamine Modules into SCID mice ......................... 41

3.3.9 Statistical Analysis................................................................................................ 42

3.4 Results........................................................................................................................... 43

3.4.1 ASC Viability........................................................................................................ 43

3.4.1.1 Addition of Laminin to Improve ASC Viability............................................... 46

3.4.2 Hydrogel Characterization .................................................................................... 47

3.4.3 ASC Differentiation .............................................................................................. 49

3.4.3.1 Oil Red O .......................................................................................................... 49

3.4.3.2 GPDH Activity.................................................................................................. 50

3.4.4 Endothelial Cell Attachment to Gels and Modules............................................... 52

3.4.5 Surface Modifications to Improve Cell Attachment............................................. 54

3.4.6 In Vivo Implantation............................................................................................. 56

3.5 Discussion..................................................................................................................... 58

3.6 Conclusions................................................................................................................... 61

3.7 Acknowledgements....................................................................................................... 61

4 Cotransplantation of ASC and HMEC in a Modular Construct Drives Early 

Angiogenesis, ASC differentiation and Long‐term Fat Regeneration in SCID Mice ..... 62

Page 7: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

vii

4.1 Abstract ......................................................................................................................... 62

4.2 Introduction................................................................................................................... 63

4.3 Materials and Methods.................................................................................................. 64

4.3.1 Cells ...................................................................................................................... 64

4.3.2 Module Fabrication............................................................................................... 65

4.3.3 Module Implants ................................................................................................... 66

4.3.4 Histology Quantification....................................................................................... 67

4.3.5 Perfusion Studies .................................................................................................. 68

4.3.6 Volume Measurements ......................................................................................... 68

4.3.7 Statistical Analysis................................................................................................ 68

4.4 Results........................................................................................................................... 69

4.4.1 Angiogenesis & Remodeling Study (Day 3, 7, 14, 21) ........................................ 69

4.4.1.1 Appearance at Explant ...................................................................................... 69

4.4.1.2 Effect of ASC on HMEC Viability and Blood Vessel Formation .................... 70

4.4.1.3 Host Tissue Infiltration and Angiogenesis........................................................ 75

4.4.2 Implants with HUVEC.......................................................................................... 77

4.4.3 Fat Development and Volume Maintenance Study (Day 30, 60, 90)................... 79

4.4.3.1 Fat Development............................................................................................... 79

4.4.3.2 Angiogenesis and HMEC Viability .................................................................. 83

4.4.3.3 Volume Measurements ..................................................................................... 85

4.5 Discussion..................................................................................................................... 86

4.6 Conclusions................................................................................................................... 89

4.7 Acknowledgements....................................................................................................... 89

5 Investigating the Relationship between Adipose‐dervied Stem Cells and 

Endothelial Cells In Vitro.................................................................................................................. 90

5.1 Abstract ......................................................................................................................... 90

5.2 Introduction................................................................................................................... 91

5.2.1 Cells ...................................................................................................................... 92

5.2.2 Optimized In Vitro Angiogenesis Sprouting Assay.............................................. 92

5.2.3 Sprouting Counts and Measurements ................................................................... 94

5.2.4 RNA Isolation ....................................................................................................... 95

Page 8: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

viii

5.2.5 cDNA Synthesis.................................................................................................... 96

5.2.6 Real-time RT PCR ................................................................................................ 97

5.2.7 Statistical Analysis.............................................................................................. 100

5.3 Results......................................................................................................................... 101

5.3.1 Effect of ASC on HUVEC Sprouting from Modules in a Fibrin Gel................. 101

5.3.2 RT-PCR Analysis ............................................................................................... 106

5.4 Discussion................................................................................................................... 111

5.5 Conclusions................................................................................................................. 114

5.6 Acknowledgements..................................................................................................... 115

6 Synopsis.......................................................................................................................................116

6.1 Biomaterials for Adipose Tissue Engineering ............................................................ 117

6.2 Revascularization Strategies ....................................................................................... 119

6.3 Understanding the relationship between ASC and EC ............................................... 122

6.4 Future Directions ........................................................................................................ 124

7 Conclusions ................................................................................................................................128

8 Reference List............................................................................................................................130

9 Appendix 1:  HMEC Harvesting............................................................................................143

Page 9: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

ix

List of Figures 

Figure 1-1: Modular approach to adipose tissue engineering....................................................... 3

Figure 2-1: Major phases in the development of mature adipocytes from multipotent stem cells

found in the stromal-vascular fraction of adipose tissue. ............................................................... 9

Figure 2-2: Poloxamine is modified with methacrylate groups (IEM) that can undergo free

radical polymerization. ................................................................................................................. 17

Figure 2-3: Mechanisms of angiogenesis. .................................................................................. 19

Figure 2-4: Functions of adipose tissue vasculature................................................................... 22

Figure 2-5: Modulators of angiogenesis in adipose tissue.......................................................... 23

Figure 3-1: Alamar Blue® viability of ASC embedded in collagen (control), poloxamine-only

and collagen/poloxamine hydrogels over 7 days .......................................................................... 44

Figure 3-2: Live/Dead™ confocal microscopy images of ASC embedded in collagen and

collagen/poloxamine hydrogels. ................................................................................................... 45

Figure 3-3: Live/Dead™ confocal microscopy images of ASC embedded collagen/poloxamine

hydrogels with the addition of laminin. ........................................................................................ 47

Figure 3-4: Immunostaining of Collagen and Collagen/Poloxamine/Laminin hydrogels.......... 48

Figure 3-5: High magnification images of immunostained collagen/poloxamine hydrogels..... 49

Figure 3-6: ASC differentiation in collagen, poloxamine and collagen/poloxamine hydrogels 50

Figure 3-7: GPDH activity of ASC during differentiation. ........................................................ 51

Figure 3-8: GPDH activity of ASC during differentiation normalized for GAPDH expression.52

Figure 3-9: HMEC-1 attachment to poloxamine hydrogel films................................................ 53

Figure 3-10: HMEC-1 attachment to poloxamine modules........................................................ 54

Figure 3-11: HMEC-1 attachment to modules with fibronectin treatment................................. 55

Figure 3-12: HMEC-1 attachment to modules with collagen coating. ....................................... 56

Figure 3-13: Collagen/Poloxamine modules implanted subcutaneously in SCID mice............. 57

Figure 4-1: Photographs of the modules at explant day 7 and 14............................................... 70

Figure 4-2: UEA-1 Lectin stained sections of HMEC+ASC modules and HMEC-only modules

at day 3, 7 and 14. ......................................................................................................................... 71

Figure 4-3: Magnified images of serial sections of the tissue surrounding a single module at day

3..................................................................................................................................................... 73

Page 10: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

x

Figure 4-4: Magnified images of serial sections of a single blood vessel surrounding a module

at day 3.......................................................................................................................................... 74

Figure 4-5: MVD counts from HMEC+ASC implants at day 3, 7 and 14. ................................ 75

Figure 4-6: CD31 images of host tissue infiltration into the modular implant at day 14. .......... 76

Figure 4-7: Magnified images of serial sections of tissue surrounding several modules. .......... 76

Figure 4-8: MicroCT images of HMEC+ASC (A) and HMEC-only (B) module implants under

mouse skin at day 21..................................................................................................................... 77

Figure 4-9: UEA-1 Lectin stained sections of HUVEC+ASC modules and HUVEC-only

modules at day 3, and 14............................................................................................................... 78

Figure 4-10: Oil Red O images of frozen sections from HMEC+ASC and ASC-only module

implants at 30, 60 and 90d post implantation. .............................................................................. 80

Figure 4-11: H&E and Masson’s Trichome micrographs of HMEC+ASC modules at day 60. 81

Figure 4-12: Masson’s Trichrome micrograph of HMEC+ASC modules at day 90.................. 81

Figure 4-13: Percentage of area stained red for Oil Red O on micrograph sections of

HMEC+ASC and ASC-only module implants at day 30, 60 and 90............................................ 82

Figure 4-14: H&E and Masson’s Trichrome micrographs of HMEC+ASC and ASC-only

modules 30 days after implantation. ............................................................................................. 84

Figure 4-15: UEA-1 stained sections of HMEC+ASC modules 30 days and 90 days after

implantation. ................................................................................................................................. 85

Figure 4-16: Volume measurements of HMEC+ASC and ASC-only module implants. ........... 86

Figure 5-1: Experimental set up of the in vitro angiogenesis assay. .......................................... 94

Figure 5-2: Image of capillary like-formations growing out from the surface of a HUVEC-

coated module in fibrin gel with the presence of ASC in coculture at day 3. .............................. 95

Figure 5-3: Photographs of HUVEC sprouting from modules in a fibrin gel with and without

ASC coculture............................................................................................................................. 103

Figure 5-4: Magnified image of HUVEC sprouting from modules with ASC coculture at day

14................................................................................................................................................. 104

Figure 5-5: Average number of HUVEC sprouts from the module surface in fibrin gel with and

without the presence of ASC in coculture. ................................................................................. 105

Figure 5-6: Average length of HUVEC sprouts from the module surface in fibrin gel with and

without the presence of ASC in coculture. ................................................................................. 106

Page 11: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

xi

Figure 5-7: Relative fold change in mRNA expression of adipogenic genes of ASC in coculture

with HUVEC, vs. ASC cultured alone........................................................................................ 108

Figure 5-8: Relative fold change in mRNA expression of angiogenic genes of ASC in coculture

with HUVEC, vs. ASC cultured alone........................................................................................ 109

Figure 5-9: Relative fold change in mRNA expression of HUVEC in coculture with ASC, vs.

HUVEC cultured alone. .............................................................................................................. 110

Figure 9-1: Light microscopy images of cells obtained from HMEC separation trials............ 145

Figure 9-2: Cytospin slides of cells from Trial 6 and 7 immunostained with vWF ................. 146

 

Page 12: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

xii

List of Tables  

Table 1: Histology stains. ........................................................................................................... 66

Table 2: RT-PCR Primer List ..................................................................................................... 99

 

Page 13: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

xiii

Abbreviations 

ASC: Adipose-derived Stem Cells

bFGF: Basic Fibroblast Growth Factor

BM-MSC: Bone Marrow-derived Mesenchymal Stem Cells

C/EBPα: CCAAT/enhancer binding protein α

CD31: Cluster of differentiation molecule 31, PECAM

DHAP: Dihydroxyacetone phosphate

EC: Endothelial Cells

ECM: Extracellular Matrix

GPDH: Glycerol-3-Phosphate Dehydrogenase

H&E: Hematoxylin and Eosin

HMEC: Human Microvascular Endothelial Cells

HMEC-1: Human Microvascular Endothelial Cell line 1

HUVEC: Human Umbilical Vein Endothelial Cells

MMP: Matrix MetalloProteinases

MSC: Mesenchymal Stem Cells

MVD: MicroVessel Density

NADH: Nicotinamide adenine dinucleotide

PAI-1: Plasminogen Activator Inhibitor 1

PDGF: Platelet Derived Growth Factor

PECAM: Platelet-Endothelial Adhesion Molecule

PPARγ: Peroxisome Proliferator-Activated Receptor γ

SCID: Severe Combined Immunodeficiency

SD: Standard Deviation

SMA: α-Smooth Muscle Actin

SMC: Smooth Muscle Cells

SVF: Stromal-Vascular Fraction

TNFα: Tumor Necrosis Factor

TUNEL: Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling

UEA-1: Ulex Europaeus Agglutinin I

Page 14: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

xiv

VEGF: Vascular Endothelial Growth Factor

vWf: Von Willebrand Factor

Page 15: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

1

1 Introduction, Hypothesis and Experimental Plan 

1.1 Clinical Impetus 

There is a critical need for soft tissue reconstruction for patients suffering from cancer,

trauma, deep burns and congenital defects. These patients suffer from contour defects due to

loss of soft tissue, mainly comprised of subcutaneous adipose tissue. In particular, the surgical

treatment of breast cancer involves lumpectomy or mastectomy which leads to abnormal

cosmesis affecting the emotional well-being of patients and may also impair function (eg. range

of motion of arms). The American Society of Plastic Surgeons cites that over 5.2 million

reconstructive procedures were performed in 2009. Yet, despite the increasing clinical demand

in reconstructive, cosmetic and correctional surgery, there remains no optimal strategy for the

regeneration and replacement of adipose tissue. The use of autologous adipose tissues for soft

tissue replacement is one clinical approach that has proven problematic due to absorption,

subsequent volume loss and fibrosis of the transplanted tissue[1,2]. Synthetic materials such as

saline and silicone implants are widely used to replace soft tissue with various levels of clinical

success. These materials have certain drawbacks such as rupture, leakage, dislocation and

biocompatibility issues. Recently, tissue engineering strategies have been applied to soft tissue

regeneration and have the potential to overcome many of the deficiencies with autologous grafts

and synthetic implants. In this approach, adipose tissue is reconstructed by combining adipose-

derived stem cells (ASC) with an appropriate substrate, typically a polymeric scaffold. Seeding

of adipose precursor cells, onto synthetic and biodegradable scaffolds has led to adipose tissue

formation in vivo. Yet, these approaches have failed to maintain a volume-stable fat pad in vivo

over the long term due to lack of rapid vascularization upon implantation leading to ischemia and

transplanted cell death. Hence, a critical challenge in engineering large portions of adipose

Page 16: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

2

tissue is early vascularization of the engineered tissue upon implantation to avoid considerable

cell death. Diffusion alone can only supply cells with oxygen and nutrients at a distance of

150μm from a blood supply[3], which would be insufficient for a three-dimensional construct of

clinical relevance. Therefore, volume loss of adipose tissue can only be avoided through early

vascularization of the engineered tissue.

1.2 Modular Tissue Engineering 

Modular tissue engineering, as developed by our group, has the potential to shift the

traditional cell + scaffold paradigm because of its inherent uniformity, scaleability and most

importantly for adipose tissue engineering, vascularity. In this approach, functional cells (here,

human adipose-derived stem cells (ASC)) are embedded in small hydrogel rods (0.5mm

diameter, 1mm length) coated with a human microvascular endothelial cell layer (HMEC).

Many modules (hundreds or thousands) can be implanted subcutaneously at the site of a soft

tissue defect to restore volume loss (Figure 1-1). The interstitial spaces created from random

packing of the modules results in endothelialized channels that remodel and connect to the host

vasculature upon implantation. With rapid vascularization of the construct, the embedded ASC

have the potential to remain viable and differentiate into mature adipocytes, resulting in the

creation of a fat pad over time.

Bovine collagen type I gel used thus far as the module material is mechanically weak and

undergoes contraction with volume reductions of up to 90% over time, perhaps making it

unsuitable as a scaffold for a volume persistent culture of adipose tissue in vivo longterm. In

order to address the mechanical limitations presented with collagen gels, semi-synthetic

collagen-containing networks were produced by photopolymerization of a Tetronic- T1107

Page 17: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

3

(poloxamine) aqueous solution. These matrices demonstrated increased stiffness while

maintaining the advantageous cell-adhesive property of collagen.

Figure 1-1: Modular approach to adipose tissue engineering.

Each module contains embedded functional cells (ASC) and is surface coated with an human microvascular endothelial cell (HMEC) layer. Many modules implanted subcutaneously at the site of a soft tissue defect can restore normal cosmesis.

Page 18: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

4

1.3 Objectives and Hypothesis 

Premise:

Modular tissue engineering is a viable platform for producing vascularized adipose tissue

in vivo.

Objectives:

1. Fabricate and optimize the modular platform for adipose tissue engineering to promote

embedded ASC viability, differentiation and attachment of surface coated endothelial cells

(EC).

Hypothesis: The semi-synthetic collagen/poloxamine hydrogel used as the module material

promotes high viability of embedded ASC and allows differentiation of these cells into mature

adipocytes, while enabling good adhesion and coverage of EC on the module surface.

2. Evaluate the regenerative potential of the modular construct to produce vascularized adipose

tissue in vivo.

Hypothesis: The endothelialized perfusion channels created by packing many modules into a

larger construct provide an immature capillary network that remodels and anastomoses with the

host vasculature upon implantation to promote transplanted ASC viability and facilitates a long-

term (> 3months) volume-persistent culture of adipose tissue in vivo.

3. Investigate ASC/EC interaction in an in vitro sprouting assay.

Hypothesis: Coculture of ASC and HUVEC (no direct cell-cell contact) promotes an angiogenic

phenotype (i.e. migration and sprouting) of surface covered HUVEC in a fibrin gel.

Page 19: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

5

1.4 Thesis Organization 

I describe the utility of the modular approach for adipose tissue engineering. Chapter 2 is

a literature review that highlights current strategies to engineer adipose tissue, as well as

approaches used to vascularize tissue engineering constructs. In chapter 3 I examine ASC cell

interaction with the semi-synthetic collagen/poloxamine hydrogel as the module material [ie. will

ASC survive and differentiate into mature adipocytes in this extracellular environment? What

modifications are necessary to the collagen/poloxamine hydrogel to make it suitable for adipose

tissue engineering?]. In chapter 4, I investigate the regenerative potential of the modular

platform for adipose tissue in vivo. [ie. will the modular approach facilitate a long-term (>3

months) volume-persistent culture of vascularized adipose tissue in vivo?]. In chapter 5, I begin

to study the relationship between ASC and EC in in vitro coculture [ie. will the coculture of ASC

and EC promote EC migration and sprouting in vitro? What key angiogenic and adipogenic

genes are influenced by ASC/EC coculture?]. Chapter 6 summarizes the work with a synopsis

and gives recommendations for future work. Chapter 7 states the overall conclusions of this

thesis.

Page 20: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

6

2 Adipose Tissue Engineering & Adipose‐derived Stem Cell/Endothelial Cell Cotransplantation as a Revascularization Strategy 

2.1 Adipose‐derived Stem Cells and Soft Tissue Engineering 

Adipose-derived stem cells (ASC) are found in the stromal vascular fraction (SVF) of

adipose tissues. These adult stromal cells are similar in phenotype and multipotency to bone

marrow derived mesenchymal stem cells (BM-MSC) and have gained much interest in recent

years because of the near limitless supply of adipose tissue and the relative ease with which it

can be harvested via liposuction or abdominoplasty procedures. Traditional soft tissue

engineering approaches combine ASC and a suitable polymeric scaffold. Once implanted in the

body, the ASC are expected to proliferate and differentiate into adipocytes, while the scaffold

remodels and degrades resulting in a mature fat pad that retains the volume needed to correct the

soft tissue defect.

Adipose-derived stem cell (ASC) is a term that has been proposed to include the entire

population of adipose precursors, ie. both non-committed and committed (preadipocyte) stem

cells found in the stromal-vascular fraction of adipose tissue. For the purposes of this work, the

terms ASC and preadipocyte are used interchangeably.

2.1.1 Adipose Tissue 

Adipose tissue is a highly specialized connective tissue that is found throughout the

human body. It can be classified into two forms: white adipose tissue (WAT) and brown adipose

tissue (BAT). While both of these forms serve to insulate and cushion the body, they have

individualized functions as well. BAT is so named because of the colour due to its highly

vascular nature and it functions primarily as a heat source for the body. Since BAT is primarily

Page 21: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

7

only found in neonates and hibernating animals, WAT is the type of fat of interest in the

development of a tissue-engineered adipose substitute for reconstructive purposes.

The primary cellular component of WAT is a collection of lipid filled cells known as

adipocytes held in place by a loose connective tissue comprising mainly collagen, laminin and

fibronectin. This extracellular matrix (ECM) interconnects adipocytes where they exists in

clusters or fat lobules in the adipose tissue. Adipocytes are rounded cells, characterized by a

single large lipid vacuole, comprising approximately 90% of the cell volume, and a flattened

eccentric nucleus[4]. Other cellular components found in adipose tissue are the stromal-vascular

cells which include endothelial cells, smooth muscle cells, fibroblasts, blood cells and adipose

progenitor cells or preadipocytes[5].

While WAT has long been recognized as an inert mass with roles in maintenance of body

contours, insulation and mechanical protection, it is now recognized as a secretory organ that

functions in energy storage and balance under tight hormonal control. WAT acts as the major

energy reserve by storing triacylglycerol in periods of energy excess and mobilization during

energy deprivation. A much more complex and dynamic role of WAT has emerged recently

with realization that adipocytes secrete factors known to play a role in immunological responses,

vascular diseases and appetite regulation[6]. Leptin, for example, is a hormone that is secreted

by mature adipocytes that interacts with the hypothalamus to control satiety. When body fat

decreases the levels of leptin also decrease which in turn leads to increase in appetite and food

intake. Conversely, when the levels of body fat increase, levels of leptin also increase which

suppresses appetite. Other hormones secreted by adipocytes include immune system related

proteins such as adipsin, acylation stimulation protein (ASP), adipocyte complement-related

protein (Acrp30/AdipoQ), macrophage inhibitory factor (MIF) and tumor necrosis factor-α

Page 22: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

8

(TNFα), and vascular function related proteins such as angiotensinogen and plasminogen

activator inhibitor type I (PAI-1). The immune system related proteins may be involved in

energy homeostasis and insulin resistance. TNFα, for example, has been shown to inhibit

differentiation of adipose precursors into adipocytes and may decrease the expression of markers

of adipogenesis[7]. TNFα may also contribute to insulin resistance by inhibition of the insulin

receptor[8]. It has been shown to be present at elevated levels in obese individuals and may act

upon adipocytes to increase secretion of the proinflammatory cytokine MIF[9]. PAI-1 is

secreted by adipose tissue and higher levels are observed in omental fat compared with

subcutaneous fat. This may be involved in vascular diseases related to an increase in abdominal

adipose tissue in obesity[10]. It is clear that adipocytes act not only as a paracrine/autocrine cell

but adipose tissue has many endocrine functions as well.

2.1.2 Adipocyte Differentiation 

Adipocytes are derived from multipotent mesenchymal stem cells. Adipogenesis, the

formation of mature, terminally-differentiated adipocytes through the differentiation of precursor

cells, can occur throughout life and is likely the primary cause of increase in body fat mass[11].

The molecular mechanisms that govern the differentiation of adipose precursors to mature

adipocytes has been reviewed in the literature[6,12-15]. The differentiation cascade may be

initiated by hormones such as insulin growth factor-1 (IGF) and insulin which have been clearly

shown to be necessary for adipogenesis[16,17]. Since it is difficult to classify distinct cellular

intermediates between stem cells and mature adipocytes, adipogenesis is generally broken down

into three distinct phases: 1. Determination or commitment; 2. Growth arrest and clonal

expansion and; 3. Terminal differentiation. An overview of these events is presented in Figure

2-1.

Page 23: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

9

Figure 2-1: Major phases in the development of mature adipocytes from multipotent stem cells found in the stromal-vascular fraction of adipose tissue.

Multipotent stem cells with the capacity to differentiate into several mesenchymal lineages give rise to the preadipocyte as a result of adipogenic inducers. The cells undergo growth arrest and clonal expansion and subsequent terminal differentiation. The cells change shape from a splindle-like fibroblastic morphology to a rounded cell with increased size while accumulating lipid droplets. The mature adipocyte is a unilocular cell. Changes in gene expression are indicated on the right with arrows suggesting their approximate duration. Reprinted by permission from Macmillan Publishers Ltd: Journal of Craniofacial Surgery [12], copyright (2007).

Determination or Commitment

The first phase, known as determination or commitment, involves the commitment of the

pluipotent stem cells to the adipocyte lineage, which results in the conversion of the multi-potent

stem cell to the preadipocyte. These precursors cells are small (diameter 10-12μm), fusiform or

fibroblast-like in appearance and do not contain any intracellular lipid[4]. The factor or gene

Page 24: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

10

responsible for the commitment of the stem cell to the adipocyte lineage is unknown. One

theory is that the signal is secreted by nearby mature adipocytes, as they have been shown to

secrete factors that promote preadipocyte proliferation and differentiation[18]. Bone

morphogenic protein 4 (BMP4) has been shown to cause 10T1/2 cells (multipotential cell line) to

commit to the adipocyte lineage at high frequency in vitro, suggesting its role in preadipocyte

committal[19]. Pre-adipocyte factor-1 (Pref-1) is another factor that has been proposed to

maintain the phenotype of the preadipocytes. Commencement of differentiation of preadipocytes

has been shown to coincide with decreased expression of pref-1 and that pref-1 is highly

expressed in 3T3 preadipocytes but not mature fat cells[20].

Growth Arrest and Clonal Expansion

Growth arrest of the proliferating phenotype of preadipocytes is the next step in the

differentiation process. It appears that growth arrest and not the actual cell-cell contact is needed

for differentiation[6]. The transcription factors, CCAAT/enhancer binding protein α (C/EBPα)

and peroxisome proliferator-activated receptor γ (PPARγ) are two key transcription factors that

control adipocyte differentiation. While their expression increases dramatically during adipocyte

differentiation, low levels of these factors expressed in preadipocytes may be responsible for

initiating growth arrest in the cells. Studies have demonstrated both the antimitotic activity of

C/EBPα[21] and the ability of PPARγ to induce cell cycle withdrawl[22]. After growth arrest,

studies on preadipocytes cell lines demonstrate that these cells undergo at least one round of

DNA replication and cell division leading to the clonal expansion of the committed cells[23].

Yet, for primary human preadipocytes clonal expansion may not be an important step as they do

not appear to require cell division to begin differentiation[24].

Page 25: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

11

One of the earliest changes in gene expression during differentiation is the increased

expression of lipoprotein lipase (LPL) which is often cited as an early marker for adipocyte

differentiation[25]. LPL is secreted by mature adipocytes and is involved in controlling lipid

accumulation[26]. LPL expression has been shown to occur spontaneously at confluence

without adipogenesis inducers and therefore may be involved in growth arrest and may not be a

part of an early differentiation step[27].

Terminal Differentiation

In the final and third phase, the preadipocytes undergo morphological and biochemical

changes where cells become rounded in shape and increase their diameter by up to 20 times by

accumulation of triacylglycerol and lipid vacuoles and become sensitive to insulin[6]. Terminal

differentiation is controlled through the expression of CCAAT/Enhancer binding proteins

(C/EBP) and peroxisome proliferator-activated receptors (PPAR). Both C/EBPα and PPARγ act

synergistically to activate the transcription of genes that produce the adipogenic phenotype[28].

The activity, protein and expression levels of mRNA of many enzymes involved in

triacylglycerol synthesis including adenosine triphosphate citrate lyase, malic enzyme, glycerol-

3-phosphate dehydrogenase (GPDH) and fatty acid synthase increase by up to 100-fold[6,29,30].

Glucose transporters are also increased[31] as well as proteins not directly associated with

triacylglycerol synthesis such as aP2, a fatty acid binding protein which is considered an

intermediate marker of adipocyte differentiation[29,32]. Other factors that are produced by the

maturing adipocytes include angiotensinogen II, adipsin, and leptin which are viewed as late

markers of differentiation[6,25,33].

Page 26: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

12

2.1.3 Adipose Tissue Extracellular Matrix 

The extracellular matrix of adipose tissue is a critical component that gives not only

mechanical support of the tissue but also provides sites for cell attachment, proliferation and

migration and signaling that controls a variety of processes including adipogenesis, angiogenesis,

and immune response. Structurally, the ECM of adipose tissue serves to connect adipocytes

where they form clusters or fat lobules in vivo. These lobules are highly vascular and therefore

also include endothelial cells and their respective ECM. Adipose tissue ECM is divided into two

different compartments, the basement membrane that surrounds individual adipocytes and the

reticular fibre network that forms a soft skeleton that supports the tissue. The basement

membrane is comprised of networks of collagen IV and laminin-8 linked together with entactin

and heparan sulfate proteoglycan. The reticular fibres are made of collagen type III connected to

fibres of collagen type I[12]. During differentiation, not only are there changes in cell

morphology where preadipocytes enlarge and acquire lipids, but also on the level of secreted

ECM as well. One of the first signs of differentiation, for example, is deposition of collagen at

the cell-ECM border and creation of the basement membrane[34]. Alterations in the adipose

tissue ECM by the differentiating preadipocytes are likely needed to change cell adhesion

properties, remodeling of cell components and cellular organization into fat lobules. Studies

with preadipocyte cell lines have shown that when undergoing differentiation these cells lose

their fibroblastic characteristic along with decreased expression of collagens type I and III[35]

and increased expression of collagen IV, laminin, entactin and glycosaminoglycans[36-38]. In

addition, differentiation of 3T3-F442A cells was shown to have decreased synthesis and

assembly of actin and fibronectin, with concurrent disappearance of fibrillar bundles of

extracellular matrix fibronectin in the pericellular space[39]. While preadipocytes have been

shown to alter secretion of ECM components during differentiation, the ECM may in turn have a

Page 27: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

13

role in regulating the differentiation process. Inhibition of collagen synthesis, for example, was

shown to prevent preadipocyte differentiation, highlighting that collagen synthesis is a

requirement for differentiation[40]. Furthermore, studies on fibronectin have shown that it has

both pro and anti-adipogenic properties[29,41].

2.1.4 Stem Cells for Adipose Tissue Engineering 

Adult stem cells, known as mesenchymal stromal cells (MSC), are mature adult cells that

are undifferentiated and found in a particular tissue or organ. These cells have the capacity for

self-renewal and are able to differentiate into the majority of specialized cell types found in the

body. In addition, they are easily isolated from bone marrow and adipose tissues and can be

expanded in vitro, making them an attractive cell source for tissue engineering. Bone marrow

derived mesenchymal stromal cells (BM-MSC) are likely the most recognized adult stem cell

used for therapeutic purposes. They are multipotent cells and can be differentiated into several

cell types from different lineages, including osteocytes, myocytes, adipocytes and neural

cells[42-44]. Similar to bone marrow, adipose tissue represents another source of mesenchymal

stromal cells. Adipose tissue is typically present in an excess and expendable quantity in the

body which makes ASC harvesting a relatively safe and easy procedure by liposuction or

abdonminoplasty. When given the proper stimuli, ASC harvested from the stromal vascular

fraction (SVF) of adipose tissue have been shown to differentiate into cells of adipogenic,

chondrogenic, myogenic and osteogenic lineages[45,46]. It is not surprising that both ASC and

BM-MSC are found to be very similar in both morphology and phenotype[47] since both adipose

tissue and bone marrow are derived from the mesenchyme. Directly comparing the phenotype

between ASC and BM-MSC shows that they are >90% identical[48]. Typical of MSC, ASC

have been shown to express high levels of stem cell surface markers CD29, CD73, CD90,

Page 28: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

14

CD105 and low levels of CD45[49]. In contrast to BM-MSC, ASC have been shown in several

reports to be CD34+ at early passages, with transient decrease in expression level with

subsequent expansion in culture[49-51]. It is still not clear whether CD34 can be used as a stem

cell marker to identify the ASC lineage. One group working with human adipose tissue found

that the population of the SVF that was positive for CD34 contained the ASC[52]. While species

differences may exist, another group studying murine adipose tissue found that the CD34

negative SVF population was enriched for ASC[53].

2.1.5 Adipose Tissue Engineering Strategies 

There have been several strategies investigated to engineer adipose tissue. Scaffold

guided tissue regeneration is the method most widely investigated. In this approach ASC are

cultured on absorbable polymeric scaffolds and implanted in vivo such that simultaneous cell

proliferation/differentiation and scaffold resorption results in a mature fat pad. An important

design criterion here is the artificial extracellular matrix (ECM) that is used as a three-

dimensional support for the transplanted cells. Not only must this ECM provide the necessary

mechanical support and desired degradation properties but must also provide the appropriate

cell-matrix interaction. ASC have been cultured successfully on a wide array of materials for

adipose tissue regeneration including synthetic materials such as biodegradable PLGA[54],

protein-coated polytetrafluroethylene[55], polyglycolic acid[56] and natural scaffolds such as

hyaluronan-based[57], collagen[58] and acellular placenta[59]. Hydrogel materials, such as

collagen[60] and alginate[61] gels may prove optimal because their mechanical properties

closely match soft tissue, providing a “feel” more like native adipose tissue. Synthetic hydrogels

have also been investigated, yet as ASC are anchorage-dependent cells[62], synthetic materials

typically need to be modified to promote cellular attachment in order to avoid cell death. A

Page 29: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

15

poly(ethylene glycol) hydrogel system was shown to support preadipocyte viability, for example,

only upon modification with the laminin-binding peptide sequence YIGSR[63], which is

consistent with research that demonstrates preadipocyte adhesion and migration on Laminin-1

via the α1β1 integrin[62]. The optimized hydrogel system that supported both viability and

proliferation also included the peptide sequence LGPA to permit polymer degradation by cell-

secreted collagenase[63]. Remodelling of the matrix by the cells may therefore be necessary to

create space to accommodate proliferation and cellular expansion of the ASC during

differentiation.

Another approach to repair soft tissue deficits involves the use of an acellular tissue

engineering device. This strategy, known as de novo adipogenesis, uses a stimulus, such as a

growth factor, to cause migration of ASC to the implant site where they will proliferate and

differentiate into adipocytes to form a local accumulation of adipose tissue. This method was

demonstrated by injections of Matrigel (gelatinous protein mixture secreted by Engelbreth-

Holm-Swarm (EHS) mouse sarcoma cells) with basic fibroblast growth factor (bFGF)[64,65].

The authors concluded that the visible fat pad that formed was the result of ASC and EC

migration to the injection site. In another example, photocured styrenated gelatin microspheres

with bFGF and insulin and insulin-like growth factor I (IGF-1) with a view to stimulating both

angiogenesis and adipogenesis[66]. They were successful in creating adipose tissue in vivo yet

de novo adipogenesis seemed limited in the quantity of fat tissue that can be created and may not

be clinically relevant for large soft tissue voids.

While many current strategies are successful in initially generating an accumulation of

adipose tissue in vivo, they fail to engineer an adipose tissue graft that will remain viable and

retain its volume over the long-term (> 3 months). In one of the few published long-term

Page 30: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

16

studies, the volume of adipose tissues engineered with ASC seeded onto a PLGA scaffold and

implanted subcutaneously into a rat was found to be maximal at 2 months, decreased by 3

months and completely resorbed by months 5-12[67]. In another study, ASC seeded onto

collagen scaffolds implanted subcutaneously into mice decreased in volume by approximately

70% after 3 weeks implantation[58]. The volume loss of adipose tissue may be the result of

insufficient mechanical strengths of the construct to withstand the compressive forces in vivo and

the lack of sufficient neovascularization upon implantation.

2.1.6 Semi‐Synthetic Collagen/Poloxamine Hydrogel 

Poloxamine is a collagen-mimetic as developed by our group[68] to overcome some of

the limitations of collagen hydrogel scaffolds, mainly weak mechanical properties and

progressive remodeling and contraction of the matrix. Matrix contraction, which can result in

volume reductions of up to 90% over time, is undesirable when engineering a soft tissue

replacement where maintenance of implant volume is critical.

Poly(ethylene oxide) and poly(propylene oxide) block copolymers (PEO-PPO) are

commonly used as materials in biomedical engineering. They have shown good

cytocompatibility with different cell types and they are non-irritating when applied topically or

subcutaneously, and produce little in vivo response. The poloxamines, also known as

Tetronic™) are different from the more common PEO-PPO-PEO triblock coplymers in that they

contain four arms and as such their higher functionality makes a superior degree of crosslinking

achievable with lower polymer concentrations. Collagen-containing poloxamine hydrogels are

produced by functionalization of the four-arm PEO-PPO block copolymer with methacrylate

groups (Figure 2-2) and subsequent UV free radical polymerization of water solutions of the

modified polymer in the presence of collagen. This forms a network of collagen suspended in

Page 31: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

17

the crosslinked poloxamine hydrogel, where the collagen imparts the cell adhesive properties to

the matrix while the poloxamine (non cell adhesive) provides the mechanical strength. The

crosslinked collagen/poloxamine hydrogel was found to be much stiffer than a pure collagen gel

with a storage (G’) and loss modulus (G’’) of 7400Pa and 1000Pa as compared with 70Pa and

10Pa respectively for collagen-only gel[68].

Figure 2-2: Poloxamine is modified with methacrylate groups (IEM) that can undergo free radical polymerization.

Page 32: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

18

2.2 Angiogenesis & Revascularization Strategies 

2.2.1 Mechanisms of Angiogenesis 

Angiogenesis is the process of new blood vessel formation and is a critical for tissue growth,

function and repair. Figure 2-3 highlights some of the pathological steps during new blood

vessel formation. Angiogenesis can occur though three methods: sprouting, bridging and

intussusception. Spouting is the most common process and begins with vessel destabilization;

the activation of the endothelial cell layer, vasodilation of the existing vessels, an increase in

vascular permeability and degradation of the basement membrane [69,70]. Largely in reponse to

nitric oxide (NO) production, vasodilation is one of the earliest steps in angiogenesis. VEGF, in

part due to the increase in NO production [71], mediates an increase in vascular permeability due

to the redistribution of intercellular adhesion molecules, including PECAM-1, and vascular

endothelial (VE)-cadherin, and alterations in cell membrane structure via induction of a series of

kinases [69,72]. Angiopoietins (Ang-1 and Ang-2) are ligands for the EC receptor Tie-2. While

Ang-1 is a natural anti-permeability factor, Ang-2 helps detach smooth muscle cells and loosens

the underlying matrix allowing EC to migrate as the inter-endothelial cell contacts are

broken[69]. Subsequently, the extravasation of plasma proteins, similarly induced by VEGF, is

required to provide a temporary support for the migration of activated EC. The activated EC

modify proteinase expression and release proteinases causing degradation of the basement

membrane. This is an important step as it will allow the EC to invade into the stroma of

neighborouring tissue. The degradation of the basement membrane is caused by the cooperation

of urokinase-plasminogen activator (uPA) and metalloproteinase (MMP) systems [70]. The

degradation of the ECM, not only provides “room” for the EC to migrate, but results in the

liberation of growth factors, including bFGF, VEGF, and insulin-like growth factor-1 (IGF-1),

Page 33: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

19

Figure 2-3: Mechanisms of angiogenesis.

Sprouting is the most common angiogenic process and begins with activiation of the endothelial cell layer and destablization of the basement membrane. Ang-2 helps detach smooth muscle cells and loosens the underlying matrix allowing EC to migrate by VEGF stimulation. Migrating EC assemble, form solid cords and subsequently acquire a lumen. These capillaries then synthesize a new basement membrane. Vessels are stabilized and mature via the recruitment of periendothelial support cells such as pericytes and smooth muscle cells (SMC) by PDGF. Adapted by permission from Macmillan Publishers Ltd: Nature Medicine [73], copyright (2000).

which would otherwise remain immobilized in the matrix [73]. These liberated growth factors

act on the activated EC as mediators of migration and proliferation.

Once the physical barriers in the ECM are dissolved, the next step in the process of

angiogenesis is the migration of proliferating EC into the interstitial space. At this stage there

are a myriad of factors that are responsible for mediating embryonic, neonatal and pathological

angiogenesis, including VEGF, angiopoietins, FGFs and their receptors while many other factors

Page 34: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

20

have been implicated in the process [73]. FGFs for example, stimulate EC growth and recruit

mesenchymal and/or inflammatory cells, producing countless angiogenic factors [74]. Platelet-

derived growth factor (PDGF) encourages microvascular sprouting of EC and recruits pericytes

and smooth muscle cells around nascent vessel sprouts [75].

As EC migrate into the ECM, they assemble, form solid cords and subsequently acquire a

lumen. This is achieved through the intercalation and thinning of the EC along with fusion of the

existing vessels to create longer and greater diameter vessels. The formation of the capillary

lumen requires the polarization of the EC and is established via cell-cell and cell-matrix

interactions. These capillaries then synthesize a new basement membrane. Vessels are

stabilized and mature via the recruitment of periendothelial support cells such as pericytes and

smooth muscle cells (SMC) [70].

Finally, upon fusion of the newly formed vessels with the preexisting vasculature, blood

flow is initiated. When sufficient neovascularization has occurred there is an upregulation of

anti-angiogenic factors, such as VEGF-R1, Ang-1, angiostatin, with downregulation of many of

the pro-angiogenic stimulators discussed. Upon completion of the angiogenic cascade, the EC

return to their quiescent state with survival measured in years.

Following a similar mechanism, angiogenesis can also occur through bridging and

intussusception. In bridging, vessels can be split into individual daughter cells through

development of transendothelial cell bridges. Branching by intussusception takes place when

there is an insertion of interstitial tissue columns into the lumen of pre-existing vessels [76].

Subsequent growth and stabilization of these colums, due in part to the ingrowth of

periendothelial cells, results in partitioning of the vessel lumen and remodeling of the vascular

network.

Page 35: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

21

2.2.1.1 Vascular Functions in Adipose Tissues 

With increasing interest in the development of a therapeutic target for obesity, the

complex interplay between the different cellular components of adipose tissue is only now being

increasingly understood and has been reviewed in [77]. Figure 2-4 shows the functional

relationship of adipose tissue to its vasculature. In growing adipose tissue, angiogenic vessels

contribute to adipogenesis through several distinct mechanisms. First and most obvious, is that

the vessels bring nutrients and oxygen to nourish the main component of the fat tissue – the

adipocytes. Like any other tissue in the body, they require sustenance for growth and survival.

Secondly, the circulating plasma provides an environment rich in growth factors and cytokines.

Leptin and adiponectin, for example, are secreted adipose-derived hormones that are responsible

for maintenance of adipose tissue mass[78]. Thirdly, the angiogenic vessels permit infiltration of

inflammatory cells, such as monocytes and neutrophils, which tend to be observed in high

numbers in obese individuals[79]. The fourth purpose of the vasculature is to supply circulating

stem cells from the bone marrow. These mesenchymal cells can differentiate into preadipocytes,

adipocytes and vascular cells, including pericytes and endothelial cells[80,81]. Fifth, a recent

study has shown that adipocytes descend from a pool of proliferating progenitor cells that reside

in the mural cell compartment of adipose vasculature, but not in the vasculature of other tissues.

Mural cells including pericytes, have been shown to be capable of differentiating into both

adipocytes and preadipocytes making the interactions between the fat and vascular compartments

even more complex[81]. The sixth functional relationship of adipose tissue vasculature is the

paracrine relationship shared between activated EC and adipocytes/preadipocytes which is

discussed in more detail in section 2.2.1.3.

Page 36: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

22

Figure 2-4: Functions of adipose tissue vasculature.

Blood vessels in adipose tissue provide transport of oxygen, nutrients, hormones, growth factors (GFs), inflammatory cells (ICs) and bone marrow derived mesenchymal stem cells (BM-MSC) that maintains the functions of adipocytes and adipose tissue. In the circulation, BM-MSCs, ICs and endothelial progenitor cells (EPCs) participate in both angiogenesis and adipogenesis. Endothelial cells in angiogenic vessels produce growth factors and adipokines. Adipocytes also produce angiogenic factors and adipokines that induce angiogenesis. Perivascular cells such as pericytes (PC) can differentiate into pre-adipocytes that become adipocytes. Reprinted by permission from Macmillan Publishers Ltd: Nature Reviews Drug Discovery [77], copyright (2010).

2.2.1.2 Angiogenic Modulators 

As seen in Figure 2-5, adipocytes and their precursors along with inflammatory and

stromal cells in adipose tissue work together as mediators in the angiogenic process. Adipocytes

secrete angiogenic adipokines and traditional angiogenic factors such as VEGF and bFGF.

Leptin, for example, has been shown to increase vascular permeability and to synergistically

stimulate angiogenesis with VEGF and bFGF[82]. Lipids secreted by fat cells may also be pro-

angiogenic. Monobutyrin, for example, has been shown to stimulate angiogenesis in vivo and

Page 37: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

23

Figure 2-5: Modulators of angiogenesis in adipose tissue.

Adipocytes and their precursors along with inflammatory and stromal cells in adipose tissue work together as mediators in the angiogenic process though secretion of multiple angiogenic factors, adipokines and cytokines. Adapted by permission from Macmillan Publishers Ltd: Nature Reviews Drug Discovery [77], copyright (2010).

microvascular endothelial cell migration in vitro[83]. Besides pro-angiogenic molecules, fat

tissue also produces several angiogenesis inhibitors including thrombospondin I and

plasminogen activator inhibitor (PAI-1). Overexpression of PAI-1 was found to have no effect

on de novo adipogenesis in a mouse model, while the blood vessel size in the fat pad was much

smaller than controls, indicating an inhibitory effect on angiogenesis[84].

In addition to the adipocytes, inflammatory cells play a large role in angiogenesis.

Chronic inflammation is consistently seen in obese individuals, which results in the infiltration of

Page 38: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

24

many leukocytes and macrophages. These cells produce several angiogenic growth factors,

including VEGF, tumor necrosis factor (TNFα), interleukin-6 (IL-6), IL-8 and

chemokines[79,85].

ASC play a major role in adipose tissue angiogenesis through the secretion of soluble

angiogenic molecules and possibly by differentiating into vascular cells that become part of the

endothelium. ASC produce multiple angiogenic factors including, VEGF, hepatocyte growth

factor, bFGF, transforming growth factor β, and chemokines[86]. In addition to the secretion of

bioactive molecules, recent research has shown that ASC have the ability to differentiate into

endothelial cells or perivascular pericytes so they may actively participate in the angiogenic

process. Populations of ASC that are CD34+ have been shown to preferentially differentiate into

endothelial cells, while those that have lost CD34 expression tend to express more pericyte

markers which may be related to the process of commitment or differentiation into a specific

lineage[87,88].

2.2.1.3 Relationship between ASC and EC 

Adipose tissue development is closely associated with angiogenesis but the molecular

interplay between adipogenesis on angiogenesis remains unclear. Research over the past decade

has demonstrated the paracrine relationship between adipose tissue and the vasculature and has

identified some of the pro-angiogenic and anti-angiogenic growth factors expressed by adipose

tissues or adipocytes cultured in vitro. These include VEGF, bFGF, Ang-1,-2, PAI-1, TGF-β

and leptin[89]. A recent study demonstrated that human preadipocytes and endothelial cells both

express the αvβ3 integrin and express and secrete PAI-1 which regulates preadipocyte and EC

migration in vitro[90]. This suggests that during development preadipocytes migrate with the

Page 39: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

25

developing capillary bed during angiogenesis, and that the production and secretion of PAI-1 by

the preadipocytes ensures local coordination of adipogenic/angiogenic process.

In another study that highlights the interplay between angiogenesis and adipogenesis, the

PPARγ pathway (master regulator of adipogenesis) was blocked in 3T3-F442A preadipocytes

(preadipocyte cell line) and found to not only inhibit differentiation of these cells but

angiogenesis in vivo as well[89]. They also investigated the role of vascular endothelial growth

factor receptor 2 (VEGFR2) signaling in angiogenesis, vessel remodeling and adipocyte

differentiation. VEGFR2 is expressed on vascular endothelial cells and its signaling is critical to

the angiogenic process and VEGF is highly expressed during adipocyte differentiation[89].

They found that inhibition of angiogenesis by a VEGFR2 blocking antibody not only reduced

angiogenesis and tissue growth, but also inhibited preadipocyte differentiation as well[89].

These findings demonstrate the reciprocal relationship between adipogenesis and angiogenesis

and suggest that inhibition of VEGF signaling can reduce adipose tissue formation in vivo.

In another well-defined study, EC/ASC cocultures were studied in vitro in a system

containing serum but no additional exogenous cytokines or ECM proteins to better understand

the mechanisms at work[91]. ASC were found to have a profound effect on EC to stimulate

angiogenesis and branching networks of cord structures. During coculture, there was an

observed increase in the ECM protein production by both the ASC and EC, increased α-smooth

muscle actin expression by ASC and increased CD31/platelet endothelial cell adhesion

molecule-1 (PECAM-1) on the surface of the EC. Overall, matrix metalloproteinase activity and

paracrine cell communications between ASC and EC in the VEGF, hepatocyte growth factor and

PDGF pathways were found to impact vascular network formation. As expected, ASC were

shown to exhibit a significantly higher angiogenic effect than smooth muscle cells or fibroblasts.

Page 40: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

26

These studies are in accordance with earlier work that showed how differentiation of

3T3-F442A stimulated EC chemotaxis in vitro and neovascularization in vivo[92]. Extracellular

matrix factors secreted by microvascular endothelial cells, derived from adipose tissue, have

shown to stimulate both preadipocyte proliferation[93] and differentiation[94] into mature fat

cells in primary culture.

Despite these observations, the exact mechanisms between adipogenesis and

angiogenesis are not well defined. It appears that a paracrine relationship exists between ASC

and EC that may be exploited when creating a vascularized adipose tissue engineering construct.

2.2.2 Revascularization Strategies 

Previous attempts at adipose tissue reconstruction using either autologous tissue or tissue

engineering strategies have commonly failed as a result of implant resorption due to ischemic

cell death. Native adipose tissue is highly vascular, with each mature fat cell in contact with at

least one capillary[95]. One of the challenges of tissue engineering is to provide adequate

nutrients and oxygen to all cells embedded within the tissue engineered construct, especially

those large 3D constructs of clinical relevance, as the diffusional limitation of approximately

150µm is imposed on nutrient and waste transfer[96]. As such, a successful adipose tissue

engineering strategy will promote rapid integration with the host vasculature upon implantation.

Revascularization and inosculation are two strategies by which a tissue engineered device can

become integrated with the host vasculature[97]. In a revascularization strategy, the host blood

vessels grow into and nourish the implanted device. This approach is limited, however, by the

slow kinetics of nutrient and waste diffusion and may only be suitable for small or thin

constructs. Delivery of angiogenic growth factors could be used to accelerate blood vessel

growth, however it is difficult to orchestrate the complex temporal and spatial cues required for

Page 41: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

27

stimulation and maturation of blood vessels by the delivery of but a few growth factors. A more

promising strategy to support cellular survival in larger matrices is inosculation, whereby a

capillary network is created within the adipose substitute and is subsequently fused with the host

vascular system upon implantation[98]. Several revascularization strategies have been

investigated in the literature, these include: growth factor delivery, biomaterial-mediated

angiogenesis, transplantation of primary endothelial cells, vascular pedicle, and stem cell

transplantation.

2.2.2.1 Growth Factor Delivery 

Many current studies of therapeutic angiogenesis deliver angiogenic growth factors either

directly into the circulation or at the target site through a single or a series of bolus injections.

This approach has been used successfully in cases of both peripheral[99] and myocardial[100]

ischemia, yet has the disadvantage of possibly producing unwanted blood vessel formation at

distant sites. In addition, the half life of most bolus injections of growth factors is less than one

hour, making repeated injections necessary to maintain the therapeutic effect.

In tissue engineering approaches, growth factors are combined with polymeric

biomaterials as a vehicle for localized and sustained release. Through chemical and/or physical

means, angiogenic growth factors such as VEGF, bFGF and PDGF can be incorporated into

tissue engineering scaffolds to promote angiogenesis upon implantation. Perhaps most widely

investigated in the adipose tissue engineering literature is the incorportation of bFGF into the

tissue engineering device. bFGF has been shown to enhance adipogenesis of mesenchymal stem

cells in both 2 and 3-dimensional in vitro cultures[101,102], and also reported as effective in

inducing angiogenesis, and regeneration of bone, cartilage and nerve[103-105]. Work by

Page 42: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

28

Thompson and colleagues showed that bFGF incorporated into a collagen type I gel produced de

novo adipogenesis accompanied by angiogenesis in mice[106]. However, their engineered

adipose tissue resorbed after 6 weeks once the collagen gel scaffold had degraded. Others have

investigated bFGF release from gelatin microspheres in Matrigel™, and found significant

adipose tissue generation, yet again volume maintenance was not achieved[64,65]. It appears

that while sustained angiogenic growth factor release is successful in initially generating

adiopose tissue in vivo, once the scaffold degrades and the growth factor delivery ceases, the

regenerated tissue diminishes.

2.2.2.2 Biomaterial‐mediated Angiogenesis 

An emerging interest in the field of tissue engineering is exploring the possibility that a

biomaterial itself can be used as an agonist of an angiogenic response. Here we can imagine

incorporating ASC with an appropriate biomaterial that will elicit an angiogenic response once

implanted in the body. In previous years, we have investigated a poly(butyl methacrylate-co-

methacrylic acid) tissue engineering scaffold that was shown to be pro-angiogenic in vivo

without the use of exogenous growth factors[107]. Hyaluronan is an example of a naturally

occurring polymer that has been suggested to have pro-angiogenic activity. It is an important

structural component in the ECM that is involved in numerous cell adhesion and migration

events and has been found to be naturally angiogenic when degraded into small fragments[108].

Sacchachitin, a natural polymer derived from fungal mycelia, is another biomaterial that has

shown pro-angiogenic potential[109]. The authors found that in a full-thickness skin graft model

a larger number of blood vessels were found in the sacchachitin covered wound compared to

cotton gauze control at day 3. Hisology revealed that there was an acute and rapid inflammatory

response with a larger number of neutrophils and lymphocytes in the sacchatitin treated wound

Page 43: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

29

compared to the control. Perhaps the invading inflammatory cells released pro-angiogenic

growth factors (ie. VEGF) which would explain why granulation tissue and angiogenesis

occurred faster in the sacchachitin treated wound as compared to the control.

2.2.2.3 Vascular Pedicle 

Another revascularization strategy is the use of a defined vascular pedicle to nourish the

adipose tissue engineering construct in vivo. In this approach a vascular pedicle is elevated and

inserted into a hollow plastic chamber and implanted subcutaneously. The chamber typically

contains a biomaterial and growth factors to accelerate de novo adipogenesis and to stimulate

angiogenic sprouting from the pedicle. Once the tissue has been generated, it is possible to

transfer the construct microsurgically to a remote site to repair a soft tissue defect. Morrison and

colleagues have developed an in vivo model of angiogenesis in rats, based on the microvascular

arteriovenous loop inside a polycarbonate chamber in the groin of a rat[110]. They were able to

show than when the chamber was combined with a collagen matrix, it spontaneously developed a

vascularized, three-dimensional, fibrous tissue mass. They further developed this model for

mice and showed that when Matrigel™ was used as the matrix inside the chamber, the chambers

filled with adipose tissue, creating a highly vascularized fat flap[111].

2.2.2.4 Cell‐Based Revascularization Strategies 

Cell based revascularization strategies have been studied in patients with various

ischemic diseases. These studies have shown that stem cells from bone marrow[112], skeletal

muscle[113], umbilical cord blood[114] and adipose tissue[115] can be used to treat patients

with conditions such as myocardial infarction, heart disease and peripheral vascular disease. It is

Page 44: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

30

believed that these progenitor cells have potential in promoting tissue revascularization and

restoring function through the release of many bioactive molecules.

Transplantation of EC as a revascularization strategy is a relatively unexplored approach.

Work in our laboratory has studied HUVEC transplanted subcutaneously into severely combined

immunodeficient mice (SCID). HUVEC were found to initially form vessel-like structures in

vivo but these tended to disappear after a few weeks after transplantation. We then explored the

implantation of rat aortic endothelial cells in an allogenic rat model. Stable chimeric vessels

were seen, yet only when the animals were given immunosuppressive drug treatment[116].

Others have also observed the fragile nature of EC transplantation. Human dermal endothelial

cells (HDMEC) seeded in Matrigel™ and implanted in a PLGA scaffold in SCID mice showed

initial growth and sprouting of donor derived vessels, but these regressed quickly by 14

days[117]. In another example, HUVEC in a Matrigel™ plug implanted in SCID mice showed a

decrease in HUVEC vessel density over time[118]. It appears that EC transplantation alone is

not sufficient to avoid apoptosis and drive the formation of mature, stable and functional vessels

long-term.

Several methods to achieve viability and stability of donor EC derived vessels have been

investigated and include transfecting an anti-apoptotic gene into the EC or cotransplantation with

a supporting cell. Bcl-2 is an anti-apoptotic gene that has been shown to extend EC survival in

vitro[119]. Pober and colleagues demonstrated that Bcl-2 transduced HUVEC produced an

increased density of HUVEC-lined microvessels that were perfused in vivo as compared with

untransduced or control-transduced HUVEC[120]. It appeared that Bcl-2 overexpression

recruited ingrowth of perivascular smooth muscle cells, which organized by day 60 into

HUVEC-lined multilayered stable-vessels. Similarly, another group looked at implants of

Page 45: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

31

collagen-fibronectin gels containing Bcl-2 transduced HUVEC and found that they produce

human EC and murine vascular smooth muscle cell chimeric vessels in SCID mice[121].

Microfil casting of the vasculature revealed perfusion of the donor derived vessels with the host

at day 60, demonstrating anastomosis and vessel functionality. Cotransplantation with a

supporting cell is another successful approach to ensure viability and stability of the donor EC

vessels. In angiogenesis, other vascular cells such as fibroblasts, smooth muscle cells and

pericytes migrate to help support and stabilize the growing vasculature. As such, it is not

surprising that cocultures of pericytes and EC have stabilized vessels in 3D collagen gels[122]

and that fibroblasts were shown to stabilize tubular network formation in tissue-engineered

reconstructed skin[123]. Perivascular precursors (10T1/2 cells) and embryonic fibrobasts have

also been co-cultured with HUVEC in PLGA scaffolds[124]. The vascular network created by

the cotransplantation of HUVEC and these precursors in SCID mice was shown to be functional

and stable for 1 year in vivo. These precursor cells likely differentiated into smooth muscle cells

which acted as a support for donor vessel maturation and stability.

In adipose tissue engineering, the use of ASC as the supporting cell in an EC

transplantation revascularization strategy has only just begun to be investigated. Increasing

evidence has shown that the ASC in adipose tissue may reside in a perivascular location and that

pericytes and endothelial cells may actually be progenies of the ASC[125]. ASC may induce

angiogenesis through paracrine support[50] of EC and suppression of their apoptosis[126,127]

and may actually integrate into the host vasculature[127]. In addition, ASC have been shown to

have pericyte-like activity in vitro by stabilizing endothelial networks[87]. Stark and colleagues

were the first to show a patent connection of tissue-engineered microvessels in adipose tissue to

a host vessel system without applying exogenous angiogenic growth factors of transfection with

Page 46: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

32

an anti-apoptotic gene[128]. They combined HDMEC spheroids and ASC in a fibrin matrix in a

chorioallantoic membrane (CAM) model. After 7 days, many vessels consisting of HDMEC

could be detected and the vessels were perfused with chick erythrocytes. In a recently published

article, ASC were combined with endothelial progenitor cells (EPC) in collagen implants and

implanted subcutaneously in SCID mice[129]. The ASC were shown to cooperate with the

EPCs to coassemble into a functional vessel network. Synergism by cotransplantation of ASC

and EPC increased the density and complexity of the vascular networks many fold greater than

in implants with ASC or EPC alone. The authors speculated that the angiogenic effect that ASC

have on EC may be the result of secretion of diffusible proangiogenic and antiapoptotic factors

(such as VEGF and Ang-1) and direct contact with forming endothelial tubes.

Since most strategies to engineer adipose tissue employ ASC, it seems that EC

cotransplantation is a promising revascularization strategy.

Page 47: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

33

3 Semi‐Synthetic Collagen/Poloxamine Hydrogel as a Potential Biomaterial for Adipose Tissue Engineering 

3.1 Abstract 

With increasing clinical demand for an adipose tissue substitute, a suitable biomaterial that

possesses the appropriate mechanical properties and ‘feel’ of fat along with the necessary

biological properties for cellular transplantation and regeneration is needed. A semi-synthetic

collagen-containing poloxamine hydrogel was investigated for its feasibility as a novel

biomaterial for adipose tissue engineering. Adipose-derived stem cells (ASC) embedded in the

photocrosslinked collagen/poloxamine hydrogel displayed lower viability over 14d in culture as

compared with ASC in collagen gels. Immunostaining of the collagen/poloxamine hydrogel

revealed inhomogeneous distribution of collagen within the hydrogel, resulting in regions devoid

of any cell attachment. Adipogenic differentiation was confirmed by Oil Red O staining in the

collagen/poloxmine hydrogel, yet was observed only in regions of the hydrogel rich in collagen.

Human microvascular endothelial cells (HMEC-1) attached and proliferated onto the surface of

both collagen/poloxamine hydrogel films and modules, yet incomplete endothelial cell coverage

was achieved over 14d in culture. The immiscibility of collagen and poloxamine made it

undesirable for a biomaterial for adipose tissue engineering.

3.2 Introduction 

Tissue engineering has the potential to address the clinical need for the repair and

regeneration of adipose tissue defects. Millions of surgeries are performed each year to restore

normal cosmesis resulting from soft tissue traumas and abnormalities, including mastectomies,

tumor removal, deep burns and congenital defects. Current treatment modalities use autologous

and allogenic adipose tissues with limited success as these therapies are associated with

Page 48: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

34

operative risk, donor site morbidity and progressive resorption/fibrosis of the implanted tissue

due to insufficient neovascularization of the fat graft[54,130].

Recent approaches to adipose tissue engineering combine Adipose-derived Stem Cells

(ASC) and a suitable biomaterial that is used as a three-dimensional support for the transplanted

cells. A biomaterial for adipose tissue engineering must provide the necessary mechanical

support and “feel” but must also provide the appropriate cell-matrix interaction for ASC viability

and differentiation. ASC have been cultured successfully on a wide array of materials for

adipose tissue regeneration including synthetic materials such as biodegradable PLGA[54],

protein-coated polytetrafluroethylene[55], polyglycolic acid[56] and natural scaffolds such as

hyaluronan[57], silk[131] and collagen scaffolds[58]. Hydrogel materials, such as collagen[60]

and alginate[61] gels may prove optimal because their mechanical properties closely match soft

tissue. Collagen has the advantage of biodegradability, low antigenicity and cell-binding

properties, yet it is mechanically weak. Upon implantation it undergoes remodeling and

contraction with volume reductions of up to 90%, making it not ideal for an application where

volume maintenance is critical[132]. Synthetic hydrogels have tunable mechanical properties,

yet may need to be modified to promote cellular attachment in order to avoid cell death. A

poly(ethylene glycol) hydrogel system, for example, was shown to support ASC viability only

upon modification with the laminin-binding peptide sequence YIGSR[63], which is consistent

with research that demonstrates ASC adhesion and migration on Laminin-1 via the α1β1

integrin[62].

A semi-synthetic collagen/poloxamine hydrogel was developed by our group[68] to

combine both the biological properties of collagen and the improved mechanical stiffness of the

synthetic hydrogel. Poly(ethylene oxide) and poly(propylene oxide) block copolymers (PEO-

Page 49: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

35

PPO) are commonly used as materials in biomedical engineering[133]. They have shown good

cytocompatibility with different cell types and they are non-irritating when applied topically or

subcutaneously, and produce little in vivo response[133]. The poloxamines, (also known as

Tetronic™) are different from the more common PEO-PPO-PEO triblock coplymers in that they

contain four arms and as such their higher functionality makes a superior degree of crosslinking

achievable with lower polymer concentrations. The crosslinked collagen/poloxamine hydrogel

was found to be much stiffer than a pure collagen gel with a storage (G’) and loss modulus (G’’)

of 7400Pa and 1000Pa as compared with 70Pa and 10Pa respectively for collagen-only gel[68].

In addition, cytocompatibility of embedded and surface-attached cells was high [134].

Mechanical strength is not the only criterion in designing a hydrogel for adipose tissue

engineering. The effective attachment of EC to the module surface and the phenotype of

embedded ASC and their differentiation into mature adipocytes must be considered. It was

thought the unique 2-phase network of collagen/poloxamine may be advantageous for ASC

differentiation. The ASC are anchorage dependent and need to attach to the matrix to proliferate

and avoid cell death by anoikis[63]. During differentiation, these cells must be able to remodel

the matrix (ie. using cell secreted collagenase) to provide room for volume expansion during

lipogenesis. Mature adipocytes, on the other hand, may prefer a non cell-adhesive matrix. It has

been suggested that a biomaterial suitable for adipose tissue engineering may need a balance

between cell adhesive qualities (attachment and spreading) and non-adhesive properties that

favour differentiation[135].

Here, we report on the feasibility of using collagen/poloxamine as a novel biomaterial for

adipose tissue engineering. Studies were performed to assess embedded ASC viability,

differentation and EC cell attachment.

Page 50: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

36

3.3 Materials and Methods 

3.3.1 Cells 

A protocol for the isolation and culture of human adipose-derived stem cells (ASC) has

been adopted from Dr. Lauren Flynn, Queen’s University[136]. Fresh, sterile abdominal fat

samples were obtained from patients undergoing elective surgery, with consent. The tissue was

minced and digested in a 2mg/mL collagenase solution at 37oC, containing Kreb’s Ringer

bicarbonate buffer, supplemented with 3mM glucose, 25mM HEPES and 20mg/mL BSA (all

Sigma, Oakville, ON). Upon digestion the sample was filtered to remove undigested tissue

fragments and centrifuged to separate the desired stromal-vascular cell population from the

buoyant mature adipocytes. The cell suspension was gently agitated in erythrocyte lysing buffer

and filtered again to remove red blood cell fragments. Several centrifugation steps with washes

in complete media finished the processing. The cells were plated in a 1:1 mixture of Dulbecco’s

Modified Eagle’s Medium and Ham’s F-12 nutrient mixture (DMEM:Ham’s F-12) (Sigma,

Oakville, ON), supplemented with 10% fetal bovine serum (FBS) (Sigma, Oakville, ON), 100

U/mL penicillin and 0.1 mg/mL streptomycin (Gibco, Burlington, Canada). The growth medium

was changed every 2-3 days. To passage the cells, cultures at 90% confluence were trypsin-

released (0.25% trypsin/0.1% EDTA, Gibco, Burlington, Canada), washed, counted and re-

plated in new flasks at 30,000 cells/cm2. Cells at passage 2 were used for the differentiation

experiments, while higher passage cells (but < passage 10) were used for the viability

experiments.

To induce adipogenic differentiation, cells were cultured in serum-free DMEM:Ham’s F-

12 supplemented with 15 mM NaHCO3, 15 mM HEPES, 33 μM biotin, 17 μM pantothenate, 10

μg/mL transferrin, 100 nM cortisol, 66 nM insulin, 1 nM triiodothyronine (T3), 100 U/mL

Page 51: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

37

penicillin and 0.1 mg/mL streptomycin. For the first 72 hours of differentiation, 0.25mM

isobutylmethylxanthine (IBMX) and 1 μg/mL of troglitazone were added to the differentiation

medium (all Sigma, Oakville, ON).

The Human Microvascular Endothelial Cell line-1 (HMEC-1) was used in the cell

attachment studies to overcome the limitations of primary cells, mainly difficulty in isolation and

limited life span. HMEC-1 is an immortalized human microvascular endothelial cell line that

has been shown to maintain the morphologic, phenotypic, and functional characteristics of

normal human microvascular endothelial cells[137].

3.3.2 Hydrogel Synthesis 

Poloxamine (T1107-methacrylate) is synthesized under nitrogen by a reaction with

Tetronic 1107 (a gift from BASF, New Jersey, NJ) and isocyanato ethylmethacrylate (IEM)

using stannous (II) 2-hexyl-hexanoate (SnOct) as the catalyst (Sigma, Oakville, ON), as

described elsewhere[68].

To prepare poloxamine hydrogels, an aliquot of T1107-methacrylate solution (25 wt% in

distilled water) was mixed with a 3mg/mL collagen type I solution (PureCol™, Inamed,

Fremont, CA) or phosphate buffered saline (to produce poloxamine hydrogels without collagen)

to the desired final concentration. Then 10X MEM (10.1 g α-Minimum Essential Medium

powder (Gibco, Invitrogen, Paisley, PA), and 2.2g endotoxin free NaHCO3, in 100mL endotoxin

free water) was added and mixed thoroughly and neutralized to pH ~ 7.4 by adding the required

volume of 0.8N NaHCO3. Irgacure I2959 ((2-hydroxy-1-[4-(2-hydroxyethoxy)phenyl]-2-methyl

propanone) (Ciba, Basel, Switzerland), dissolved in ethanol, was added so that the final

concentration of photoinitiator is 0.07wt %. The poloxamine solution was then mixed with the

desired cell number (ie. 106 cells/mL) and transferred into the wells of a 96-well plate (to make

Page 52: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

38

hydrogel films) or injected into the lumen of PE tubing (which is subsequently cut after

crosslinking to produce cylindrical modules). The poloxamine-cell solutions were incubated at

37oC for 30min (to promote collagen fibrillogenesis) and then crosslinked by exposure to UV

light for 10min (Spectroline EN-180 lamp, one 8-W 365nm tube, with intensity of

1.850mW/cm2(at source)) at 10cm (distance from source UV). Hydrogel films (or modules)

were washed repeatedly upon crosslinking with complete media and subsequently cultured at

37oC. Collagen-only gels were prepared in the same manner, only by omitting the poloxamine-

methacrylate.

In an attempt to improve cell attachment, hydrogels of collagen/poloxamine/laminin were

prepared by adding laminin (derived from Engelbreth-Holm-Swarm murine sarcoma basement

membrane in 1 mg/mL in Tris buffered NaCl, Sigma) with the collagen/poloxamine solution (5

μg/mL and 50μg/mL laminin) prior to mixing with cells and photocrosslinking

3.3.3 Cell Viability 

A preliminary alamarBlue® viability assay (Invitrogen) (106 ASC/mL hydrogel, 10min

UV, 0.07wt% I2959) comparing collagen, poloxamine-only and collagen/poloxamine hydrogels

was conducted to assess the viability of the ASC in culture over 7 days. AlamarBlue® contains

a redox indicator that changes color in response to metabolic activity. Briefly, alamarBlue®

reagent was added directly into culture media of the gels in a 96-well plate, after 1, 3, and 7 days

after hydrogel fabrication, to a final concentration of 10% and the plate was returned to the

incubator. Optical density was measured at 540 and 630 nm with a standard spectrophotometer

after 4 hours of incubation at 37oC. The percent reduction was calculated and compared against

ASC embedded in collagen-only gels.

Page 53: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

39

A Live/Dead® kit (Invitrogen) was used to stain live and dead ASC cells within the

crosslinked hydrogel. Confocal microscopy was used to visualize cells and estimate viability by

observing the presence of green (live) and red (dead) staining cells.

3.3.4 Hydrogel Characterization 

Immunostaining of collagen and laminin was used to visualize the distribution of the

components in the hydrogels. Briefly, the monoclonal anti-collagen type I produced in mouse

(Sigma, Oakville, ON) and anti-laminin produced in rabbit were diluted 1:100 in PBS and 100uL

of this solution was added to each well and incubated for 10min. After several PBS washes the

secondary antibodies, Alexa Fluor® goat anti-mouse 568 (red) and Alexa Fluor® goat anti-rabbit

488 (green) (Molecular Probes) were diluted 1:1000 in PBS and added to the gels and incubated

for 10min. After several PBS washes, the hydrogels were visualized under confocal microscopy.

3.3.5 Oil Red O 

Lipid accumulation within the collagen, poloxamine, collagen/poloxamine and

collagen/poloxamine/laminin hydrogels was assessed by Oil Red O (Sigma, Oakville, ON)

staining at 7 and 14 and 21 days after the induction of differentiation. At each time point, three

hydrogels from each group were fixed for 24 hours in 10% neutral buffered formalin and rinsed

thoroughly with PBS. The hydrogels were labelled prior to Oil Red O staining with the anti-

collagen type I and Alexa Fluor® 588 carboxylic acid, succinimidyl ester, as described in section

3.3.4 to visualize the distribution of collagen in the hydrogel. Hoescht (Invitrogen) was added at

a concentration of 1µg/mL and allowed to incubate at room temperature for 10min followed by

extensive PBS rinsing.

An Oil Red O stock solution (3 g/L) was prepared in isopropanol and was diluted 3:2

(v:v) with deionized water. This solution was agitated at room temperature for 10 minutes and

Page 54: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

40

then filtered through a type 1 qualitative filter paper. Each hydrogel was incubated in 0.5 mL of

the prepared stain for 2 hours under agitation at room temperature. All samples were rinsed

thoroughly overnight in deionized water, with three solution changes to reduce non-specific

staining. The scaffolds were analyzed using confocal microscopy (Zeiss LSM510, C-APO

63x/1.2 NA water immersion (DIC) objective lens, excitation with a HeNe laser at 543 nm and

an Enterprise laser at 351 nm).

3.3.6 Glycerol‐3‐phosphate Dehydrogenase Activity 

Glycerol-3-phosphate dehydrogenase activity (GPDH, a mid to late marker of ASC

differentiation) was measured for each hydrogel configuration at 72h, 7d and 14d post induction

of differentiation using a GPDH activity measurement kit (Kamiya Biomedical Co.) to assess the

impact of each type of hydrogel on the differentiation of ASC (n=4). The kit provides a substrate

reagent containing NADH and DHAP that is mixed with the prepared samples. The decrease in

the NADH concentration, as DHAP is converted into glycerol-3-phosphate by the GPDH in the

sample, can be measured by monitoring the change in the absorbance of the mixture at 340nm.

A larger change in the absorbance during the analysis period correlates with greater GPDH

activity in the sample. Using the extinction coefficient of NADH, it is possible to calculate the

GPDH (Units/mL) in the sample. To allow comparison of the different samples (varying cell

viabilities) the results were normalized for cell number as determined by semi-quantitative

Western blotting for the housekeeping gene GAPDH.

3.3.7 EC Attachment to Gels and Modules 

To characterize endothelial cell attachment to the collagen/poloxamine hydrogel, both

hydrogel films (100μL in wells of a 96-well plate) and modules were prepared as outlined in

section 3.2.2. To the hydrogel films, 30,000 HMEC-1 were seeded on top of the films and

Page 55: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

41

cultured in ‘coculture media’ (DMEM-F12 Ham’s, 10% FBS, 1% P/S, 10μg/mL hydrocortisone,

1ng/mL EGF, 10μg/mL L-glutamine) (Sigma, Oakville, ON). Modules were dynamically and

then statically seeded with HMEC-1 (2x107 HMEC-1/mL of settled modules) by first rocking the

modules in 10mL of cell suspension for 1h at 37oC and subsequently allowing the cells to settle

and attach to the modules overnight in a petri dish. At 1, 7 and 14d post-seeding, films or

modules were rinsed in PBS and stained using the Live/Dead® (Molecular Probes) viability

stain, which stains live cells green and dead cells red, and visualized with confocal microscopy

(Zeiss LSM510).

In an attempt to improve HMEC-1 attachment, several modifications including

fibronectin and collagen coatings on the surface of the modules were tested. Fibronectin (FN)

was selected as it is typically used as an adsorbed layer on the surface of tissue-culture surfaces

to promote attachment, proliferation and spreading of a variety of cell types via the Arg-Gly-Asp

(RGD) domain. Collagen/poloxamine modules were suspended in a fibronectin solution (500ug

FN per mL of settled modules, BD Biosciences) at 37oC for 1h to allow adsorption. To coate the

modules with a layer of collagen, collagen/poloxamine modules were suspended in a collagen

solution (3mg/mL, 1.5mg/mL or 0.75mg/mL bovine collagen type I (PureCol)) and neutralized

to pH 7.4 with NaHCO3. Modules were strained from excess collagen solution on a 100μm

mesh filter and the collagen coating allowed to gel at 37oC for 1h.

3.3.8 Implantation of Collagen/Poloxamine Modules into SCID mice 

Adult male (6 weeks of age) male SCID mice were purchased from Charles River

Laboratories (Wilmington, MA). They were individually housed in sterile cages and fed ad

libitum under the approval by the University of Toronto animal care committee. As a pilot

study, ~ 250 modules suspended in PBS were injected subcutaneously on the dorsum of mice.

Page 56: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

42

After 3 days, the mice were euthanized, and the implants excised and fixed in 4% neutral

buffered formalin (Sigma Aldrich, Oakville, ON) and fixed for 48h. Tissue samples were

embedded in paraffin and sections were processed and stained for hematoxylin and eosin (H&E,

Fisher) and Masson Trichrome (Fisher). Histology was performed by the Research Pathology

lab at Toronto General Hospital.

3.3.9 Statistical Analysis 

All statistical analyses were performed using the software program STATISTICA Version

5.1 (Statsoft, USA). Student’s t-tests were used to compare the means between two groups

analyzed and considered significant at p<0.05.

Page 57: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

43

3.4 Results 

3.4.1 ASC Viability 

The first step in demonstrating the efficacy of a poloxamine/collagen hydrogel system for

adipose tissue engineering was to investigate the viability of embedded ASC. A preliminary

alamarBlue® (AB) viability assay comparing collagen, poloxamine-only and

collagen/poloxamine hydrogels was conducted to assess the viability of the ASC in culture over

7 days (Figure 3-1). It was observed that cells in poloxamine and poloxamine/collagen

hydrogels had similar AB reductions over the first 3 days in culture as compared to the collagen

controls (with and without UV/initiator), suggesting that the fabrication procedure

(photocrosslinking) had little impact on cell viability. After 7 days, however, a 50% loss in the

AB reduction in the poloxamine-only hydrogel suggested cell death, likely the result of lack of

cell attachment to the matrix and death by anoikis. The viability of cells in the

collagen/poloxamine hydrogels was higher in comparison with the poloxamine-only hydrogel at

7d, yet lower when compared with collagen controls at day 7.

Page 58: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

44

0

10

20

30

40

50

60

Control (collagen) Poloxamine Poloxamine + Collagen

% A

B re

duct

ion

1d

3d

7d

Figure 3-1: Alamar Blue® viability of ASC embedded in collagen (control), poloxamine-only and collagen/poloxamine hydrogels over 7 days

Low viabilities in the poloxamine-only hydrogel after 7 days was likely due to lack of cell attachment to the matrix. ASC in collagen/poloxamine hydrogels had improved viabilities compared with poloxamine-only hydrogels at day 7, but not as good as collagen-only control (n=3). Error bars represent 1SD.

A Live/Dead™ viability stain was used to visualize the viability of the embedded ASC

over 7 days in collagen and collagen/poloxamine hydrogels (Figure 3-2). High viability was

observed in both the collagen control and collagen/poloxamine hydrogels at day 1 indicating that

the fabrication and photocrosslinking process was likely non-toxic to cells. After 7 days

however, ASC in the collagen gels spread, adopted a typical fibroblast-like morphology and

appeared to have proliferated in the matrix. In the collagen/poloxamine hydrogel at day 7,

however, pockets of viable cells similar in appearance to those found in the collagen controls

were observed, but there were also regions of the hydrogel with a high number of dead cells.

This was thought to have been the result of inhomogeneity in the distribution of the collagen and

Page 59: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

45

poloxamine phases in the hydrogel. Regions in the matrix rich in collagen would permit ASC

adherence and proliferation, while regions in the hydrogel composed of mostly poloxamine

would result in cell death over 7 days.

Figure 3-2: Live/Dead™ confocal microscopy images of ASC embedded in collagen and collagen/poloxamine hydrogels.

High viability of cells in both hydrogels at day 1 show that the fabrication process is non-toxic to cells. Two images of collagen/poloxamine at 7d show different regions of high and low cell viability in the same gel. This suggests inhomogeneity in the mixing of the collagen and poloxamine components (live cells = green, dead cells = red, scale bar = 100µm). Images are typical of 4 replicates.

Collagen

Collagen/ Poloxamine

1d 7d

Page 60: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

46

3.4.1.1 Addition of Laminin to Improve ASC Viability 

Several groups have investigated including laminin-derived peptide sequences into their

synthetic polymers to promote the attachment and proliferation of ASC[63,138]. Laminin (LN)

is a major component in adipose extracellular matrix and has been shown to promote the

adhesion, migration of ASC and may play an important role in adipogenesis[62]. It was of

interest to incorporate laminin into the collagen/poloxamine hydrogels to see if this had any

effect on ASC viability. Hydrogels of Collagen/Poloxamine/Laminin revealed similar results to

those obtained with collagen/poloxamine matrices (Figure 3-3). High cell viability throughout

the matrix was observed at day 1, with regions of viable and non-viable cells seen at day 7.

Interestingly, the addition of laminin appeared to have an effect on cell attachment. Even at day

1, cells in the hydrogel containing 50μg/mL LN appeared to be adopting a fibroblast-like

morphology and spreading onto the matrix. This was clearly seen for both the hydrogels

containing 5μg/mL and 50μg/mL laminin at day 7.

Page 61: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

47

Figure 3-3: Live/Dead™ confocal microscopy images of ASC embedded collagen/poloxamine hydrogels with the addition of laminin.

ASC in the hydrogels with laminin appear to adopt a more elongated and fibroblast-like morphology indicative of adhesion and spreading onto the matrix (live cells = green, dead cells = red, scale bar = 100µm). Images are typical of 4 replicates.

3.4.2 Hydrogel Characterization 

Immunostaining of collagen and laminin was used to visualize the distribution of the

components in the hydrogels to help explain the observed pockets of live and dead cells in the

matrix after 7 days (Figure 3-4). Collagen is stained red, laminin green and poloxamine is not

stained and appears black. Large black regions void of biological components are observed in

the collagen/poloxamine/laminin hydrogels. Laminin association with collagen is apparent at

higher magnification as both red and green colours are colocalized. Higher magnification

1d

7d

0µg/mL LN 50µg/mL LN 5µg/mL LN

Page 62: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

48

images of collagne/poloxamine show that black regions devoid of biological components span

diameters greater than 100µm (Figure 3-5).

Figure 3-4: Immunostaining of Collagen and Collagen/Poloxamine/Laminin hydrogels.

Immunostaining enables visualization of the distribution of components in the collagen/poloxamine/laminin hydrogel. Collagen is stained red, laminin green. Poloxamine is unstained and appears black. Images are typical of 4 replicates.

Collagen

Collagen/ Poloxamine/Laminin

Page 63: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

49

Figure 3-5: High magnification images of immunostained collagen/poloxamine hydrogels.

Higher magnification images of immunostaining of collagen/poloxamine hydrogels shows collagen (red) and large areas of poloxamine (black) greater than 100µm in diameter. Images are typical of 4 replicates.

3.4.3 ASC Differentiation 

3.4.3.1 Oil Red O 

The next step in demonstrating the efficacy of a poloxamine/collagen hydrogel system for

adipose tissue engineering was to investigate the differentiation capacity of the embedded ASC.

Three hydrogel configurations were tested: poloxamine, collagen/poloxamine and

collagen/poloxamine/laminin (50μg/mL LN) and compared against ASC on TCPS as a control.

As seen in Figure 3-6, in the poloxamine (POL) hydrogels lipid accumulation (red) was seen in

the majority of cells after 7 days of differentiation. However, lipid accumulation appeared to

halt after 7 days with no further increase in lipid droplet size over time and only very few nuclei

appeared to stain for Hoescht (blue) suggesting that most of these cells had died after 21 days.

Collagen/poloxamine (COL POL) and collagen/poloxamine/laminin (COL POL LN) hydrogels

diplayed intracellular lipid accumulation at day 7, with an increase in the size of the fat droplets

Page 64: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

50

at day 21 indicating a more-differentiated phenotype. Differentiating cells were found to cluster

in regions rich in collagen within the hydrogels (green by immunostaining), and only very few

cells stained positive for hoescht (blue) outside of these regions, similar to what is seen in POL

hydrogels. No observable difference was seen in the hydrogels with laminin compared to those

without. These results indicate that poloxamine hydrogels did not support ASC differentiation,

likely because of cell death due to anoikis since this hydrogel did not support cell attachment.

Collagen/poloxamine hydrogels appeared to support ASC differentiation with levels similar to

that of collagen gels, yet only in regions of the hydrogel rich in collagen.

Figure 3-6: ASC differentiation in collagen, poloxamine and collagen/poloxamine hydrogels

Oil Red O staining (red) of ASC embedded in collagen (COL) poloxamine (POL), collagen/poloxamine (COL POL) and collagen/poloxamine/laminin (COL POL LN) hydrogels cultured with adipogenic medium at 7 and 21 days. Hoescht stains cell nuclei blue. Immunostaining of collagen is green. Scale bar = 20μm. Images are typical of 4-6 replicates.

3.4.3.2 GPDH Activity 

The overall GPDH expression for each sample is shown in Figure 3-7 with the data

normalized for total number of cells in Figure 3-7. ASC in all samples appear to be expressing

COL POL COL POL COL POL LN

7d

21d

Page 65: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

51

GPDH at all time points investigated with the ASC in collagen gel samples at 72h expressing a

statistically significant higher amount of total GPDH compared to cells on TCPS or the other

hydrogel configurations. When normalized for the total number of cells, cells in the collagen

gels at 72h and 7d expressed a statistically significant lower amount of GPDH (ie. GPDH per

cell) compared to all other samples.

Overall GPDH

0

5

10

15

20

25

TCPS COL POL COL POL COL POL LN

mU

72h7d14d

Figure 3-7: GPDH activity of ASC during differentiation. GPDH activity in mU per sample (not normalized for cell number) of each hydrogel group at 72h, 7d and 14d after the induction of differentiation (n=4). Collagen gels expressed a significantly higher total amount of GPDH than other samples at 72h (p<0.05). Error bars represent 1 SD.

*

Page 66: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

52

Overall GPDH (normalized with GAPDH)

0

24

68

1012

1416

18

TCPS COL POL COL POL COL POL LN

mU

/GA

PDH

72h7d14d

Figure 3-8: GPDH activity of ASC during differentiation normalized for GAPDH expression.

GPDH activity in mU per sample, normalized for the number of viable cells by GAPDH expression (western blot), of each hydrogel group at 72h, 7d and 14d after the induction of differentiation (n=4). Collagen gels expressed a significantly lower amount of GPDH than other samples at 72h when normalized for total cell number (p<0.05). Error bars represent 1 standard deviation.

3.4.4 Endothelial Cell Attachment to Gels and Modules 

As expected, HMEC-1 on either TCPS or collagen gel films (COL) quickly formed a

confluent layer of cells covering the surface of the films (Figure 3-9). HMEC-1 on the surface of

the poloxamine-only hydrogel (POL) appeared in rounded clusters on day 1, indicative of a non-

adhesive substrate and very few cells were found to be attached to the poloxamine-only hydrogel

film by day 14. The incorporation of collagen in the collagen/poloxamine (COL POL)

hydrogels greatly improved the attachment of the HMEC-1, yet complete coverage of the entire

surface of the film was not achieved. These holes or gaps in the surface coverage of the cells

were also observed in the collagen/poloxamine hydrogels with laminin (50μL/mL LN) (COL

POL LN). No improvement in HMEC-1 attachment to the hydrogels with laminin was observed,

likely because laminin self-associates with the collagen phase in the gels. Similar results were

found for the attachment of HMEC-1 to the surface of the collagen/poloxamine modules (Figure

* *

Page 67: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

53

3-10). Here the gaps in the surface coverage appear larger, likely due to the curved geometry of

the cylindrical module.

The lack of complete HMEC-1 coverage on the surface of the collagen/poloxamine

hydrogel was consistent with the non-uniform distribution of collagen and poloxamine within the

hydrogel (Figure 3-5). Regions that are devoid of HMEC-1 are likely regions of the gel rich in

non-cell adhesive poloxamine, while regions that are rich in collagen promote HMEC-1

attachment.

Figure 3-9: HMEC-1 attachment to poloxamine hydrogel films.

Live/Dead™ images show attachment of HMEC-1 to collagen (COL), poloxamine-only (POL), collagen/poloxamine (COL POL) and collagen/poloxamine with laminin (COL POL LN) hydrogels. Lack of HMEC-1 surface attachment is clearly observed on the POL hydrogel. HMEC-1 attachment was improved with the COL POL hydrogel yet complete surface coverage is not achieved after 14 days as compared to HMEC-1 cultured on collagen gels. The addition of laminin did not appear to improve HMEC-1 attachment. Images are typical of 4-6 replicates.

TCPS POL COL POL COL POL LN

1d

7d

COL

14d

Page 68: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

54

Figure 3-10: HMEC-1 attachment to poloxamine modules.

Live/Dead™ images show attachment of HMEC-1 to collagen/poloxamine (COL POL) and collagen/poloxamine with laminin (COL POL LN) modules. Holes or gaps in HMEC-1 coverage are likely the result of inhomgeneous distribution of the collagen/poloxamine components. Images are typical of 4 replicates.

3.4.5 Surface Modifications to Improve Cell Attachment 

Live/Dead™ images of HMEC-1 seeded onto the FN-modified modules display a similar

pattern of attachment as unmodified collagen/poloxamine modules, with incomplete coverage of

the module surface after 14 days (Figure 3-11). Again, due to the inhomogeneous mixing of the

collagen/poloxamine components, regions of the module surface rich in poloxamine (PEG-based

material) likely resisted protein adsorption and as such did not aid in promoting universal

HMEC-1 attachment over the entire surface.

1d 7d 14d

COL POL

COLPOLLN

Page 69: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

55

Figure 3-11: HMEC-1 attachment to modules with fibronectin treatment.

Live/Dead™ images show attachment of HMEC-1 to collagen/poloxamine modules modified with surface adsorbed FN show no improvement in HMEC-1 attachment. Regions of the module surface rich in poloxamine likely resisted FN adsorption. Images are typical of 4 replicates.

To overcome the issue with lack of protein adsorption to regions of the surface rich in

poloxamine, the next strategy employed was coating the modules in a collagen solution, and

gelling the collagen to create a layer of collagen gel surrounding individual modules.

Live/Dead® images of the HMEC-1 seeded onto collagen-coated modules demonstrated very

high initial attachment at day 1 for all the collagen concentrations tested (Figure 3-12). Yet, over

time the attached cells appeared to proliferate and contract the surrounding collagen gel, pulling

it off the surface of the modules, since the surrounding collagen gel was not attached well to the

module surface. This appeared as highly fluorescent regions on the surface of the modules that

seemed to detach from the module surface at day 7 and 14.

1d 7d 14d

Page 70: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

56

Figure 3-12: HMEC-1 attachment to modules with collagen coating.

Live/Dead™ images show attachment of HMEC-1 to collagen/poloxamine modules coated with different concentrations of collagen gel. Initial attachment of the cells was good (day 1) but over time proliferating cells were found to detach from the modules. Images are typical of 4 replicates.

3.4.6 In Vivo Implantation 

As a pilot study to test the feasibility of exploring the collagen/poloxamine hydrogel in

vivo, we implanted modules made from collagen/poloxamine into SCID mice using the same

model for the implantations as seen in Chapter 4. A large infiltration of neutrophils was

observed in collagen/poloxamine module implants at day 3 suggesting a highly inflammatory

reaction in vivo (Figure 3-13). This was the first implantation of poloxamine in vivo and a

biocompatibility study is needed. Perhaps additional washing steps are required to remove

residual chemicals from the synthesis steps or initiator from UV photocrosslinking.

0.75mg/mL 3mg/mL 1.5mg/mL

1d

7d

14d

Page 71: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

57

Figure 3-13: Collagen/Poloxamine modules implanted subcutaneously in SCID mice.

H&E and Masson’s Trichrome photomicrographs of collagen/poloxamine modules at day 3. Many neutrophils are surrounding the modules suggesting a large inflammatory reaction (n=3). Scale bar 100µm.

H&E Trichrome

Page 72: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

58

3.5 Discussion 

There is a need for a suitable biomaterial for adipose tissue engineering that has the

appropriate mechanical properties combined with bioactive cues that will enable ASC

attachment and differentiation into mature adipocytes, with the goal of creating a volume-stable

fat pad. Hydrogels based on synthetic polymers are advantageous in that they can be tailored

based on their physicochemical properties to suit a specific application. Hydrogels made from

poly(ethylene oxide) and poly(proplylene oxide) block copolymers (PEO-PPO) have been

extensively explored as a biomaterial because of their intrinsic biocompatibility, low protein

binding and hydrophilicity[133]. Yet, these properties make cell attachment difficult. Here we

report on the feasibility of using a semi-synthetic collagen/poloxamine hydrogel for adipose

tissue engineering. This network consists of 2 components; first the poloxamine provides the

crosslinks to form the hydrogel while collagen is a non-binding element that provides improved

biological properties.

We found that the collagen/poloxamine is not a suitable biomaterial for adipose tissue

engineering, at least without further modification. While embedded ASC viability was higher in

collagen/poloxamine hydrogel as compared to the poloxamine-only case, it was not as high as

the collagen-only control. Even with the addition of laminin in an attempt to improve embedded

ASC viability, Live/Dead® staining revealed pockets of both viable and dead cells throughout the

collagen/poloxamine hydrogel. With regions greater than 100µm in diameter devoid of any

biological components, the heterogeneous nature is likely the cause of the regions of non-viable

cells observed after 7 days; these cells were likely in a region rich in poloxamine and were

unable to attach to the PEO-rich matrix. The immiscibility of the collagen and poloxamine and

the resultant inhomogeneous distribution of the components within the hydrogel was likely the

Page 73: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

59

result of entropy-driven phase separation between poloxamine and collagen. A similar problem

was encountered in a recent report of a Pluronic F-127 hydrogel for adipose tissue engineering

where collagen was added to promote cell attachment and spreading. Even the addition of a very

small amount of collagen resulted in a single cluster of collagen and cells within the matrix[139].

Oil Red O staining revealed extensive lipid accumulation in the collagen-only hydrogels,

as expected. Some lipid accumulation was observed by cells in the poloxamine-only hydrogel at

day 7, yet very few cells stained positive for Hoescht, suggesting that most of the embedded cells

had apoptosed by this time. Oil red O staining was extensively found in the collagen/poloxamine

and collagen/poloxamine/laminin hydrogels, but only in the regions that also stained positive for

collagen. No observable improvement was seen in the hydrogels with laminin, likely since the

laminin and collagen were colocalized. It appears that initial ASC attachment to the matrix is

critical for cell viability, and differentiation.

Glycerol phosphate dehydrogenase (GPDH) activity demonstrated some level of ASC

differentiation within all hydrogels tested, with collagen hydrogels giving the highest amount of

GPDH per sample, likely due to the highest number of viable cells. When the GPDH activity

was normalized for cell number, interestingly collagen gels at 72h and 7d expressed a

statistically significant lower amount of GPDH (ie. GPDH per cell) compared to all other

samples. This may be explained by the fact that ASC in the collagen gels were readily

proliferating at the onset of differentiation, while cells on TCPS had reached confluence and

cells in the poloxamine based materials are less likely to proliferate due to the non cell adhesive

nature of the gel. Cells at confluence or which are unable to proliferate due to lack of cell

attachment have reached a state of terminal differentiation which is a prerequisite for

adipogenesis. This has been seen in studies examining the effect of cell density and cell shape

Page 74: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

60

on lineage commitment of mesenchymal stem cells (MSC). MSC were seen only to differentiate

into adipocytes if they were plated on a small island of fibronectin which forces them into an

unspread and contracted morphology[140]. While the total GPDH activity from the collagen

gels may be higher, the actual fraction of cells undergoing differentiation may be lower than that

of ASC on TCPS or in poloxamine based hydrogels since many ASC were still likely

proliferating. This again hints to the possibility that an optimized biomaterial for adipose tissue

engineering will combine both a cell adhesive nature for initial attachment, spreading and

proliferation and a non-adhesive property to enhance ASC differentiation.

In the case of modular tissue engineering, not only is the viability and differentiation of the

embedded ASC important, but the biomaterial must enable EC attachment to the surface. We

found that HMEC-1 were able to attach and proliferate onto both the surface of

collagen/poloxamine hydrogel films and modules yet complete EC coverage was not achieved

over 14 days in culture. Again, the inhomogeneity of the semi-interpenetraing network likely

resulted in regions of the hydrogel surface devoid of any biological components (ie. collagen or

laminin). Modification of the module surface with either fibronectin or collagen gel coating did

not appear to be successful in creating a confluent surface layer of EC. Complete EC surface

coverage may not be critical for in vivo applications if we think of the modules simply as a

delivery vehicle for cell transplantation. Perhaps transplanted EC will remodel into vessels in

vivo regardless of whether or not they are they are in a quiescent confluent layer on the module

surface. As long as a sufficient number of EC have been transplanted this may be enough for the

creation of a vascular network in vivo. However, the inability of the collagen/poloxamine

hydrogel to support high ASC viability and differentiation makes it unsuitable for applications in

adipose tissue engineering. Perhaps, a better approach would be to chemically modify the

Page 75: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

61

poloxamine to contain bioactive groups (e.g. YIGSR) on the polymer backbone to improve

cellular compatibility with the synthetic hydrogel.

3.6 Conclusions 

The collagen/poloxamine hydrogel is not a suitable biomaterial for adipose tissue

engineering. The inhomogeneous nature of the hydrogel resulted in low viability and

differentiation of embedded ASC and incomplete EC coverage on the surface. Furthermore an in

vivo study showed that collagen/poloxamine modules caused a large inflammatory reaction. It

appears that further chemical modification of poloxamine is needed to make the hydrogel more

cell-compatible. Since this is not within the scope of this project, collagen gel was used as the

module material when studying the effect of the ASC/HMEC interaction in vivo since it avoided

any biomaterial-related complications.

3.7 Acknowledgements 

The authors acknowledge the financial support by NSERC, the gift of Tetronic 1107 from

BASF and Dr. Alejandro Sosnik for his help and guidance with polymer synthesis.

Page 76: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

62

4 Cotransplantation of ASC and HMEC in a Modular Construct Drives Early Angiogenesis, ASC differentiation and Long‐term Fat Regeneration in SCID Mice 

4.1 Abstract 

A modular approach to adipose tissue engineering was explored by embedding adipose-

derived stem cells (ASC) in sub-mm sized collagen rods or ‘modules’ and coating with

microvascular endothelial cells (HMEC). When hundreds of these modules were injected

subcutaneously into a SCID mouse, HMEC on modules containing embedded ASC appeared to

detach from the modules to form vessels as early as day 3 as confirmed by the human EC

specific UEA-1 lectin stain, and these vessels persisted for up to 90 days. Vessels numbers

decreased over 14 days, but vessel size increased suggesting a maturing of the vasculature.

Vessel perfusion with the host was confirmed at 21 days by microCT. HMEC on modules

without embedded ASC remained attached to the module surface at day 3 and UEA-1 staining

disappeared over 14 suggesting cell death. It appeared that cotransplantation with ASC had an

anti-apoptotic and angiogenic effect on HMEC. The early revascularization strategy was

successful in supporting ASC viability and differentiation, as progressive fat accumulation in the

HMEC+ASC implants was observed: ~60% of the implant area stained positive for Oil Red O

by day 90. ASC-embedded modules without HMEC surface coating did not show fat

accumulation within the implant. All implant volumes decreased over the time-course of the

experiment, yet HMEC+ASC module implants were larger than ASC-only implants at day 90.

Collagen gel is mechanically weak and contracts in vivo making it unsuitable as a biomaterial for

adipose tissue engineering where volume maintenance is critical. When combined with an

appropriate biomaterial, the modular approach to adipose tissue engineering may represent a

successful strategy to engineer soft tissue substitutes of clinical relevance.

Page 77: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

63

4.2 Introduction 

In a typical approach, adipose tissue is reconstructed by combining adipose-derived stem

cells (ASC) with an appropriate substrate, typically a polymeric scaffold. Seeding of adipose

precursor cells, onto synthetic (ie. PLGA) and biodegradable scaffolds (ie. collagen) has led to

adipose tissue formation in vivo[54,58,67]. While many of these approaches to adipose tissue

engineering have been successful in initially generating adipose tissue in vivo, they have failed to

maintain the volume of the regenerated tissue over a long-implantation period (> 3 months)[141].

Native adipose tissue is highly vascular, with resting values of blood flow and capillary filtration

coefficients two to three times higher than those in skeletal muscle. As a result, a critical

challenge in engineering large portions of adipose tissue is early vascularization of the

engineered tissue upon implantation to avoid considerable cell death. A successful adipose

tissue engineering strategy will promote angiogenesis soon after implantation to facilitate

transplanted cell survival.

Here, functional cells (adipose-derived stem cells (ASC)) were embedded in small

collagen hydrogel rods (0.5mm diameter, 1mm length) coated with a microvascular endothelial

cell layer (HMEC). Implanting many modules together is capable of filling a soft tissue defect

of any geometry, with the idea that the endothelialized lining of the modules will quickly

remodel into vessels and anastomose with the host vasculature upon implantation to promote

embedded ASC viability and differentiation into fat.

The aim of the Angiogenesis & Remodeling study (day 3, 7, 14, 21) was to study the fate

of the endothelialized lining of the ASC-embedded modules to see if they have the potential to

remodel rapidly and to anastamose with the host vasculature upon implantation to avoid

ischemic conditions. In the Fat Development & Volume Maintenance study (day 30, 60, 90),

Page 78: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

64

longer time points will be investigated to determine if the modular approach to adipose tissue

engineering can produce volume-stable adipose tissue in vivo.

4.3 Materials and Methods 

4.3.1 Cells 

Primary human adipose-derived stem cells (ASC) and human microvascular endothelial

cells (HMEC) were harvested from sterile abdominal fat samples were obtained from patients

undergoing elective surgery, with consent. This procedure is based on the protocol for the

isolation and culture of human adipose-derived stem cells (ASC) from Dr. Lauren Flynn,

Queen’s University as described in section 3.3.1 with modifications to include magnetic bead

separation for HMEC. The tissue was minced and digested in a 2mg/mL collagenase solution at

37oC, containing Kreb’s Ringer bicarbonate buffer, supplemented with 3mM glucose, 25mM

HEPES and 20mg/mL BSA (all Sigma, Oakville, ON). Upon digestion the sample was filtered

to remove undigested tissue fragments and centrifuged to separate the desired stromal-vascular

cell population from the buoyant mature adipocytes. The cell suspension was gently agitated in

erythrocyte lysing buffer and filtered again to remove red blood cell fragments. Several

centrifugation steps with washes in complete media finished the processing. Half of the resultant

cell pellet was plated for ASC culture in a 1:1 mixture of Dulbecco’s Modified Eagle’s Medium

and Ham’s F-12 nutrient mixture (DMEM:Ham’s F-12) (Sigma, Oakville, ON), supplemented

with 10% fetal bovine serum (FBS) (Sigma, Oakville, ON), 100 U/mL penicillin and 0.1 mg/mL

streptomycin (Gibco, Burlington, Canada), while the other half was plated in MCDB-131 (VEC

Technologies) for HMEC purification. HMEC were purified from contaminating ASC by CD31

positive selection using MiniMacs™ magnetic bead separation columns (Miltenyi Biotech,

Page 79: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

65

Auburn, CA). The conditions for successful HMEC separation are outlined in Appendix 1.

Primary Human Umbilical Vein Endothelial Cells (HUVEC, Lonza, Walkersville, MD), cultured

in EGM-2 culture medium (Lonza), were also used in one set of implantations to compare

different types of EC.

The growth medium on the cells was changed every 2-3 days. To passage the cells

cultures at 90% confluence were trypsin-released (0.25% trypsin/0.1% EDTA, Gibco,

Burlington, Canada), washed, counted and re-plated in new flasks at 30,000 cells/cm2. ASC at

passage 2-3 and HMEC at passage 4-6 were used for in vivo implantations. In the one

experiment using HUVEC, these cells were used at passage 4.

4.3.2 Module Fabrication 

To prepare modules, bovine Type 1 collagen (3mg/mL, PureCol™, Inamed, Fremont,

CA) was mixed with 10X MEM (10.1 g α-Minimum Essential Medium powder (Gibco,

Invitrogen, Paisley, PA), and neutralized to pH ~ 7.4 by adding the required volume of 0.8N

NaHCO3. ASC (106 cells/mL) were added to the collagen solution to prepare ASC-embedded

modules. This solution was cast into the inner diameter of sterile 0.71mm ID polyethylene

tubing (PE 60, Intramedic – BD Canada, Oakville, ON) and subsequently gelled for 60 minutes

at 37oC[142]. The tubing was cut into small pieces (~ 2mm long x 0.6mm diameter) using a

custom automatic tube cutter and collected in DMEM:F12 Ham’s media, where the modules

were separated from the tubing by vortexing and cultured for 72h at 37oC in a petri dish to allow

contraction of the modules by ASC. 1mL of settled modules (produced using 3m of tubing)

with or without embedded ASC, were seeded dynamically with HMEC or HUVEC (2 x 106) for

45min on a low speed shaker and incubated overnight in a 50/50 mixture of DMEM:F12 Ham

and EGM-2. ASC-only modules were prepared as above without endothelial cell seeding.

Page 80: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

66

4.3.3 Module Implants 

Adult male (6 weeks of age) male SCID/Bg mice were purchased from Charles River

Laboratories (Wilmington, MA). They were individually housed in sterile cages and fed ad

libitum under the approval by the University of Toronto animal care committee. Three different

treatment groups were studied; 1. Modules coated with EC, without embedded ASC; 2. Modules

embedded with ASC and coated with EC; 3. Modules with embedded ASC, without EC.

Approximately 250 modules suspended in PBS were injected subcutaneously using an 18G

needle on the dorsum of mice. The mice were euthanized at various timepoints (3, 7, 14, 21, 30,

60, 90days) and the implants excised and either fixed in 4% neutral buffered formalin (Sigma

Aldrich, Oakville, ON) for 48h or placed in Tissue-Tek® O.C.T. compound and flash-frozen in

liquid nitrogen for frozen tissue processing. Sections were processed and stained for

hematoxylin and eosin (H&E, Fisher) and Masson Trichrome (Fisher) and various antibodies

outlined in Table 1. Histology was performed by the Pathology Research laboratory at Toronto

General Hospital. All sections were viewed with a Zeiss Axiovert light microscope equipped

with a CCD camera.

Table 1: Histology stains.

Stain Preparation Objectives Hematoxylin & Eosin (H&E) Masson’s Trichrome

Paraffin-embedded Implant Integration Cellular Infiltration Morphology Inflammatory Response Fibrous capsule formation Locate modules

UEA-1 Lectin Paraffin-embedded Stain transplanted HMEC CD31 Paraffin-embedded Stain host (mouse)

vasculature

Oil Red O Frozen Stain fat

Page 81: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

67

4.3.4 Histology Quantification 

UEA-1 stained positive vessels at day 3, 7 and 14 were counted using a Microvessel

Density Count (MVD) method adapted from the tumor angiogenesis literature[143]. At low

power (2.5X Objective lens), the number and diameter of UEA-1 stained vessels in the implants

made of HMEC coated modules with and without embedded ASC were quantified using the

ImageJ (version 1.4.3) software program provided by the National Institutes of Health. At 2.5x

objective lens the entire section of an implant could typically be visualized and the total number

of vessels per objective field (or section) was counted (not hotspots). A total of 3 sections were

counted to give an average vessel count per implant. A total of 8 animals (8 implants) were

counted for each treatment with a total of three separate experiments conducted (n=8, N=3). The

presence of a patent lumen or presence of erythrocytes was not a prerequisite for the definition of

a microvessel, yet at this low magnification only larger vessel-like structures could be counted.

MVD counts were expressed as the number of vessels per mm2 of implant.

The level of fat development in implants made from ASC-embedded modules with and

without HMEC surface coating at 30, 60 and 90d was quantified using ImageJ. Briefly, low

magnification images (2.5X objective) of Oil Red O histological sections from 3 different

animals (3 sections per implant, n=3) at each time point and treatment group were taken. Again

at this low magnification the total area of the section containing the module implant could be

visualized. The area that was stained red by Oil Red O was calculated by the software and was

divided by the total area of the implant. The result was given as a % Area of the Implant stained

positive for Oil Red O.

Page 82: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

68

4.3.5 Perfusion Studies 

The level of vascular perfusion in the module implants was studied by microCT analysis

at 21 days. Following a published protocol[144], mice were first heparinized (100 units, LEO

Pharma Inc.) by subcutaneous injection for 5 minutes before the procedure. Animals were then

anesthetized with isofluorane and the descending aorta was cannulated and heparinized PBS

(5U/mL) was perfused at a constant pressure of 100 mmHg to flush away blood from the

vascular system. The mice were then perfused with 10mL of Microfil® solution (MV-122,

Flow-tech, Carver, MA) and the solution was allowed to polymerize for 2 hours. Implants were

excised into 4% formalin (Sigma Aldrich), embedded into 1% agar and visualized with a General

Electric Medical Systems MS8 microCT.

4.3.6 Volume Measurements 

To assess implant volume maintenance over time, for the 30, 60 and 90d implants

precisely 0.2mL (40mm3) of settled modules in PBS was injected into each implant site. At

explant, the dimensions of each implant was carefully measured with calipers and the volume

estimated using the volume equation of an ellipsoid.

4.3.7 Statistical Analysis 

All statistical analyses were performed using the software program STATISTICA Version

5.1 (Statsoft, USA). Student’s t-tests were used to compare the means between two groups

analyzed and considered significant at p<0.05.

Page 83: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

69

4.4 Results  

4.4.1 Angiogenesis & Remodeling Study (Day 3, 7, 14, 21) 

In the Angiogenesis and Remodeling Study the focus was to study the effect of embedded

ASC on the viability and angiogenic potential of the surface coated HMEC when implanted

subcutaneously in SCID mice at early time points (3, 7, 14, 21d). Implants containing modules

made with both surface coated HMEC and embedded ASC (HMEC + ASC) were compared

against implants made from modules without embedded ASC but with surface HMEC (HMEC-

only) and modules with only embedded ASC (ASC-only) as the controls. The results presented

here demonstrate a strong synergistic angiogenic response between the embedded ASC and

surface seeded HMEC.

4.4.1.1 Appearance at Explant 

Upon visual inspection at explant, the HMEC+ASC module clump appeared to be red

and vascularized by day 7 and 14 with what appeared to be blood vessels throughout the volume

of the implant (Figure 4-1). In contrast, the HMEC-only and ASC-only controls appeared to be

opaque and avascular.

Page 84: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

70

Figure 4-1: Photographs of the modules at explant day 7 and 14.

The HMEC+ASC explant appears red, with blood vessels visible throughout the module clump. Both the HMEC-only and ASC-only explants appear opaque and avascular.

4.4.1.2 Effect of ASC on HMEC Viability and Blood Vessel Formation 

One of the goals of this experiment was to show that the embedded ASC may improve

viability of transplanted HMEC and also to see if the embedded ASC would elicit an angiogenic

response from the HMEC. Both of these were observed. As seen in Figure 4-2 , the HMEC

surrounding the modules containing ASC were found to detach from the module surface and

organize into vessels. At day 3, the vessels appeared to be disorganized and immature, but by

day 7 and 14 they appear to be organized, mature and large (diameter 10-40µm) with many

containing erythrocytes suggesting anastomosis with the host. This is in contrast to the

HMEC+ASC HMEC-only ASC-only

7d

14d

Page 85: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

71

Figure 4-2: UEA-1 Lectin stained sections of HMEC+ASC modules and HMEC-only modules at day 3, 7 and 14.

The presence of ASC causes the HMEC to detach from the module surface to form vessels. HMEC on modules without ASC remain on the module surface and the UEA-1 staining diminishes over time suggesting cell death. Arrows highlight some large UEA-1 stained vessels containing erythrocytes, with some modules highlighted by dashed lines (n=8). Scale bar 100µm.

HMEC+ASC HMEC-only

3d

7d

14d

Page 86: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

72

HMEC-only control where the HMEC would remain on the module surface and staining would

diminish by day 7. By day 14 virtually no UEA-1 staining could be found in the control case

suggesting HMEC death without the presence of the ASC.

Higher magnification of serial sections of the HMEC+ASC modules revealed blood

vessels surrounding modules positive for UEA-1 containing erythrocytes as early as day 3

suggesting rapid angiogenesis and anastomosis upon implantation (Figure 4-3). These blood

vessels containing erythrocytes were positive for UEA-1 and negative for mouse CD31

confirming that they originate from the implanted HMEC (Figure 4-4).

Page 87: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

73

Figure 4-3: Magnified images of serial sections of the tissue surrounding a single module at day 3.

UEA-1+ blood vessels containing erythrocytes are observed surrounding the module, suggesting anastomosis as early as day 3 (n=8). The location of the module is highlighted by a dashed line. Scale bar 100µm.

H&E Trichrome

UEA-1

Page 88: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

74

Figure 4-4: Magnified images of serial sections of a single blood vessel surrounding a module at day 3.

The vessel stains positive for UEA-1 but not CD31 proving human origin. CD31 successfully stains other host derived vessels surrounding the implant (bottom right panel) (n=8). Arrow points to location of vessel. Scale bar 100µm.

MVD counts (Figure 4-5) from HMEC+ASC implants show that initially a large number

(>150 UEA-1+ vessels) are present in the area of the implant. Most of these vessels appeared

unorganized and are small (~50µm in diameter). By day 14, the total number of UEA-1+ vessels

had diminished considerably but the vessels that remained were large (~200µm in diameter).

These results suggest a typical angiogenic response that begins with a large number of small

immature vessels that organize into larger functional vessels.

H&E UEA-1

CD31 (mouse)

CD31 (mouse)

Page 89: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

75

Figure 4-5: MVD counts from HMEC+ASC implants at day 3, 7 and 14.

A large number of small vessels are observed at early time points, while by day 14 the number of vessels has decreased the average vessel diameter is larger. Error bars represent ±1SD. (n=8, N=3).

4.4.1.3 Host Tissue Infiltration and Angiogenesis 

As expected, it appeared that the ASC also had an effect on the angiogenic response of

the host tissue to the implanted modules. Host tissue found infiltrating the HMEC+ASC

modules was rich in CD31 positive blood vessels (Figure 4-6), while tissue invading the HMEC-

only module clump contained only a minimal number of host derived vessels. As can be seen in

Figure 4-7, both HMEC (UEA-1+) and host derived (CD31+) blood vessels surround the

modules by day 14. The embedded ASC appear to both stimulate the transplanted HMEC to

rapidly form vessels and to anastamose with the host at very early time points (day 3) while

promoting host vessel formation by day 14. The former will help avoid ischemic conditions

upon implantation while the latter will further nourish the engineered tissue during

differentiation and remodeling into a fat pad.

Page 90: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

76

Figure 4-6: CD31 images of host tissue infiltration into the modular implant at day 14. Many CD31+ blood vessels are seen in the HMEC+ASC module implant by day 14 showing host tissue infiltration. Dashed lines highlight some modules (n=8). Scale bar 100µm.

Figure 4-7: Magnified images of serial sections of tissue surrounding several modules.

Both UEA-1+ (arrows) and CD31+ (stars) blood vessels are observed surrounding the modules at day 14. Dashed line highlights module location (n=8). Scale bar 100µm.

HMEC+ASC HMEC-only

UEA-1 CD31

Trichrome H&E

Page 91: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

77

Perfusion of the module implants at day 21 was confirmed by microCT imaging (Figure

4-8). The results show an image of the implant (region marked by dashed line) under the mouse

skin. In the HMEC+ASC sample blood vessels can be seen branching off from a large vessel in

the skin infiltrating the region of the implant. Two large pools of microfil were seen within the

implant and this may be the result of leaky vessels or vessel rupture caused due to high pressure

during perfusion. The HMEC-only sample remained avascular with only blood vessels in the

skin visible. Microfil perfusion does not distinguish between host and donor derived vessels and

the perfusion seen here may be a combination of both HMEC-derived and mouse vasculature.

Figure 4-8: MicroCT images of HMEC+ASC (A) and HMEC-only (B) module implants under mouse skin at day 21. Many blood vessels can be seen infiltrating the HMEC+ASC implant (A), while the HMEC-only implant (B) remains avascular (n=2). Dashed lines show region of implant.

4.4.2 Implants with HUVEC 

With the success of the implants with the primary HMEC we were curious to see if the

ASC would elicit the same angiogenic response with another EC cell type. As with the HMEC,

HUVEC were seen to quickly detach from the modules containing ASC and form vessels, while

A B

Page 92: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

78

HUVEC on modules without ASC were found to remain on the module surface with a decrease

in UEA-1 staining over time suggesting cell death (Figure 4-9). In contrast with the HMEC

experiments, some small HUVEC vessels could be observed by day 14 in the case without ASC.

It appears the ASC accelerate the angiogenic response and increase vessels by both size and

number. We conclude that the angiogenic response seen in the experiments with HMEC was not

a special case (ie. due to both ASC and HMEC being harvested from the same donor) and that

ASC have an angiogenic effect on different types of EC on modules in vivo.

Figure 4-9: UEA-1 Lectin stained sections of HUVEC+ASC modules and HUVEC-only modules at day 3, and 14. Similar to the HMEC, ASC causes HUVEC to detach from the module surface to form vessels. HUVEC on modules without ASC remain on the module surface and the UEA-1 staining diminishes over time suggesting cell death. Unlike HMEC, some small HUVEC vessels are present surrounding modules without ASC at day 14. Some modules highlighted by dashed lines. Arrows highlight some large UEA-1 stained vessels containing erythrocytes (n=4). Scale bar 100µm.

ASC + HUVEC HUVEC-only

3d

HUVEC+ASC

14d

Page 93: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

79

4.4.3 Fat Development and Volume Maintenance Study (Day 30, 60, 90) 

Many approaches to adipose tissue engineering in the literature have been successful in

initially generating adipose tissue in vivo, but have failed to maintain the volume of the

regenerated tissue over a long-implantation period (> 3 months). We hypothesized that the early

angiogenesis from the cotransplantation of ASC and HMEC in the modules will nourish the

transplanted tissue to promote ASC differentiation into mature fat and limit resorption over time

to maintain implant volume. In this in vivo study we focused on:

1) Characterizing fat development in HMEC+ASC and ASC-only modules over longer

time periods (30, 60 and 90 days).

2) Characterizing the persistence of HMEC-derived blood vessels over time.

3) Comparing the volume measurements of the HMEC+ASC and ASC-only modules at

explant to monitor volume maintenance.

4.4.3.1 Fat Development 

Oil Red O staining on frozen sections of the modules at explant revealed progressive fat

accumulation in the HMEC+ASC modules over the 30, 60 and 90 days (Figure 4-10). By 30

days, no fat was stained in either the HMEC+ASC or ASC-only control case. By 60 days, much

red staining was seen in the region surrounding the HMEC+ASC module implant with pockets

of red staining within the module clump suggesting fat accumulation. This is confirmed with

higher magnification images of H&E and Masson’s Trichrome slides which show clusters of fat

cells within individual modules (Figure 4-11). This suggests that at least some of the fat

accumulation seen here is attributed to the transplanted ASC differentiating into mature fat cells.

In the ASC-only case a few small pockets of fat could be observed by day 60, but much less than

Page 94: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

80

Figure 4-10: Oil Red O images of frozen sections from HMEC+ASC and ASC-only module implants at 30, 60 and 90d post implantation.

Progressive fat accumulation can be observed by the increase in red staining in the HMEC+ASC module implants over time. Little to no Oil Red O staining was seen in the ASC-only case (n=3). Scale bar = 100μm.

30d

90d

60d

HMEC+ASC ASC-only

Page 95: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

81

what was observed in the HMEC+ASC case. By the 90 day time point, extensive Oil Red O

staining was seen throughout the region of the HMEC+ASC module clump indicating high level

of fat differentiation and/or accumulation. Again, Masson’s Trichrome images confirm this fat

accumulation where extensive clustering of adipose cells is seen within the implant (Figure

4-12).

Figure 4-11: H&E and Masson’s Trichome micrographs of HMEC+ASC modules at day 60.

Arrows highlight clusters of adipose cells within the individual HMEC+ASC modules (n=5). Scale bar = 100μm.

Figure 4-12: Masson’s Trichrome micrograph of HMEC+ASC modules at day 90.

The left panel shows a low magnification image of the implant (highlighted by a dashed line). Scale bar = 250μm. The right panel represents the boxed area and shows extensive fat accumulation within the implant reagion (n=3). Scale bar = 100μm.

H&E Trichrome

Page 96: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

82

The level of fat accumulation within the implants was quantified by analyzing the Oil

Red O stained sections with ImageJ to give a percentage of the total area of the implant stained

positive by Oil Red O (Figure 4-13). HMEC+ASC implants were shown to have progressive fat

accumulation in the implant region, growing to an average of 60% of the implant area stained

positive for Oil Red O by day 90. Implants made of modules with embedded ASC, but without

surface HMEC showed little Oil Red O staining at all time points investigated. It appears that

early vascularization, possibly driven by HMEC cotransplantation, is necessary for fat

development at later time points.

0102030405060708090

100

30d 60d 90d

% Im

plan

t Are

a O

il Re

d O

HMEC+ASCASC-only

Figure 4-13: Percentage of area stained red for Oil Red O on micrograph sections of HMEC+ASC and ASC-only module implants at day 30, 60 and 90.

Progressive fat accumulation is seen in the module implants containing both ASC and HMEC, while little fat accumulation is observed in ASC-embedded modules without HMEC (n=3). Error bars represent ± 1 SD.

Page 97: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

83

4.4.3.2 Angiogenesis and HMEC Viability 

A clear difference in the level of vascularization between the HMEC+ASC and ASC-only

modules was observed. Similar to the earlier time points, HMEC+ASC explants appeared to be

red and vascularized, while the ASC-only explants appeared to be opaque and avascular.

Histology confirmed these observations with the HMEC+ASC explants rich in large blood

vessels filled with erythrocytes while the ASC-only explants remained largely avascular (Figure

4-14). UEA-1 staining revealed that many of these large blood vessels were derived from the

implanted HMEC (Figure 4-15). HMEC-dervied vessels were shown to persist in the

HMEC+ASC modules as long as 90 days post-implantation (Figure 4-15).

Page 98: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

84

Figure 4-14: H&E and Masson’s Trichrome micrographs of HMEC+ASC and ASC-only modules 30 days after implantation.

Many large erythrocyte-filled blood vessels are clearly visible in the HMEC+ASC case where the ASC-only case appears avascular (n=5). Scale bar = 100μm.

HMEC+ASC ASC-only

Page 99: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

85

Figure 4-15: UEA-1 stained sections of HMEC+ASC modules 30 days and 90 days after implantation.

Large HMEC-derived vessels (arrows) are seen to persist up to 90 days in HMEC+ASC module implants (n=3-5). Scale bar = 100μm.

4.4.3.3 Volume Measurements 

Volume measurements of the explants revealed progressive shrinkage of both the

HMEC+ASC and ASC-only module implants over 30, 60 and 90 days (initial volume at

implantation = 40mm3) (Figure 4-16). This is not surprising as collagen gel used as the module

material is known to contract and degrade over time in vivo. However, surprisingly at day 90,

HMEC+ASC implants were shown to be larger on average than the ASC-only implants (p<0.05).

Perhaps, fat accumulation in HMEC+ASC helped to maintain implant volume.

30 d 90 d

Page 100: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

86

Figure 4-16: Volume measurements of HMEC+ASC and ASC-only module implants.

At explant, both HMEC+ASC and ASC-only module implants show progressive shrinkage over 90 days. At day 90, HMEC+ASC implants are statistically larger than ASC-only implants which may be attributed to the observed fat accumulation in HMEC+ASC implants. Initial volume at implantation = 40mm3. (n=3) Error bars represent 1SD.

4.5 Discussion 

The successful correction of soft tissue defects represents a challenge in plastic surgery.

Autologous tissue transfers and tissue engineering approaches to make adipose tissue substitutes

have largely been unsuccessful in maintaining a stable volume of adipose tissue in vivo long-

term (>3 months). Necrosis due to insufficient blood supply after cell or tissue transplantation

results in considerable volume loss. This is because in the first few days after transplantation,

transplanted cells are only nourished by diffusion from the surrounding tissue, which is only

sufficient at distances less than 150µm. Host derived vessels may take over 7 days to invade the

implant and may only then reach the periphery of a large 3-D construct of clinical relevance.

Volume loss of the adipose tissue construct is only avoided through early vascularization of the

transplanted tissue. The goal of the cotransplantation of HMEC and ASC in the modular

approach was to rapidly create a vascular network upon implantation, that would circumvent

0

5

10

15

20

25

30

30D 60D 90D

Volu

me

mm

3

HMEC+ASC

ASC-only*

*p<0.05

Page 101: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

87

ischemic conditions to promote ASC viability and differentiation and hence a long-term volume-

stable culture of adipose tissue in vivo.

HMEC transplantation as a revascularization strategy was successful in the implants with

modules containing ASC as a supporting cell. ASC are known to secrete many bioactive

molecules, such as vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF)

and granulocyte macrophage colony-stimulating factor (GM-CSF) which have been shown to

promote EC survival and proliferation[91]. It is not surprising that the presence of the ASC

appeared to have both a protective (promoted HMEC viability) and angiogenic effect on the

transplanted HMEC. As early as day 3, HMEC were found to detach from the surface of the

modules and organize themselves into primitive vessels with lumens and many containing

erythrocytes (Figure 4-2, Figure 4-3), suggesting rapid anastomosis with the host vasculature.

By day 7 and 14, the number of vessels had reduced but the vessels that remained were large

(~200µm diameter) suggesting that the initial leaky vessels had matured and organized during

the angiogenic process. This may be the result of secreted angiogenic factors in ASC/EC

paracrine signaling events, or perhaps some ASC differentiated into pericyte-like cells that

stabilized the maturing vasculature. This will be further discussed in Chapter 5. To confirm that

HMEC were not unique in their revascularization potential, modules made with HUVEC

displayed similar results. Successful inosculation, at least by day 21, of the HMEC+ASC

implants with the host vasculature was confirmed by MicroCT (Figure 4-8). Furthermore,

vessels derived from the transplanted HMEC (with embedded ASC) were shown to persist in

vivo for up to 90 days without drugs or the addition of anti-apoptotic genes. HMEC on modules

without embedded ASC, on the other hand, were found to remain on the surface of the modules

at day 3 and UEA-1 lectin staining was shown to disappear over 14 days suggesting cell death

Page 102: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

88

without the presence of a supporting cell (Figure 4-2). A TUNEL stain would have been useful

here to confirm apoptosis of HMEC in the absence of ASC cotransplantation.

We hypothesized that a successful revascularization strategy would promote embedded

ASC viability, differentiation and long-term persistent culture of adipose tissue in vivo. We

found that implants of modules containing both embedded ASC and surface coated HMEC

started displaying Oil Red O staining indicative of fat accumulation at day 30 post implantation

(Figure 4-10). The fat accumulation progressed until the end point of the study (90 days), where

the average Oil Red O micrograph was stained ~60% of total area for Oil Red O (Figure 4-13).

While we cannot be certain that all of this fat accumulation is derived from the transplanted

ASC, higher magnification images of H&E and Masson’s Trichrome stained sections show the

unilocular appearance of fat cells within individual modules (Figure 4-14), suggesting that at

least some of the transplanted ASC had differentiated into mature adipocytes. Interestingly,

ASC-embedded modules without the surface coating of HMEC were not shown to accumulate

fat by Oil Red O staining (Figure 4-10). Perhaps, these cells had died early on in the course of

the study due to lack of early vascularization as seen in the case with HMEC+ASC

cotransplantation. Or perhaps, EC/ASC paracrine signaling as discussed further in Chapter 5

promoted ASC differentiation.

While we were successful in demonstrating the ability of cotransplantation of HMEC and

ASC in a modular platform to create adipose tissue in vivo, we were not able to show that these

were volume-stable tissues. Volume measurements of the explants showed progressive

shrinkage of both HMEC+ASC and ASC-only implants over the time-course of the experiment.

The initial volume at implantation was 40mm3. The decrease in implant size at 30 days by more

than 50% the original volume is likely attributed to contraction of the collagen gel modules by

Page 103: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

89

both the embedded ASC and surface coated HMEC as these modules were not fully pre-

contracted in vitro prior to implantation. Although we did find that the HMEC+ASC implants to

be statistically larger than the ASC-only implants at day 90, yet these still had reduced in size by

approximately 50% from day 30 likely due to collagen degradation. A 50% reduction in the size

of an adipose tissue repair strategy is not clinically acceptable. This is the result of using

collagen gel as the module material which is known to shrink and degrade in vivo. The

development of a suitable biomaterial is still needed for adipose tissue engineering.

4.6 Conclusions 

Cotransplantation of ASC and HMEC in collagen gel modules implanted subcutaneously

into SCID mice was successful in rapidly creating a vascular network upon implantation. With

the presence of ASC, HMEC detached from the surface of the modules to form vessels that

contained erythrocytes as early as day 3. HMEC-derived vessels were shown to decrease in

number but increase in size over the first 14 days suggesting vessel maturation and persisted for

up to 90 days in vivo. With successful early vascularization, HMEC+ASC modules supported

ASC viability and differentiation to create a fat pad that persisted up to 90 days.

4.7 Acknowledgements 

The authors acknowledge the financial support by NSERC. Special thanks to Chuen Lo and

his expertise in animal surgeries. Also, thanks to Lisa Yu (Dr. R. M. Henkleman) for microCT

imaging.

Page 104: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

90

5 Investigating the Relationship between Adipose‐dervied Stem Cells and Endothelial Cells In Vitro 

5.1 Abstract 

There exists a paracrine relationship between ASC and EC, but the exact mechanisms

behind this interaction are unknown. To better understand the mechanisms of ASC-EC

interaction in the context of collagen modules we modified an in vitro angiogenesis assay to

study the sprouting of HUVEC from the surface of modules in a fibrin gel, with and without the

presence of an ASC-embedded collagen gel in a transwell insert. Although without direct cell-

cell contact, the presence of ASC in the wells promoted an angiogenic phenotype in the HUVEC.

HUVEC were seen to sprout from the modules and branch to form a dense vascular network that

intersected with vessels from neighboring modules. In the case without ASC coculture, HUVEC

were initially seen to sprout from the surface of the modules at day 3 and 7, however the average

number and length of sprouts were much less than those observed in the coculture case. By day

14, the vascular network originally created had degenerated into individual cells. It is clear that a

support cell, such as ASC, is necessary for HUVEC vascular network formation in the in vitro

conditions investigated here. The preliminary RT-PCR results were quite variable and the

presence of HUVEC did not greatly affect the mRNA expression level of adipogenic genes in the

ASC. The mRNA level of several angiogenic genes, including VEGF, PAI-1 and TNFα, were

shown to be increased in ASC with HUVEC coculture at early time points. These factors may be

involved in ASC-EC paracrine signaling.

Page 105: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

91

5.2 Introduction 

Creation of a functional vascular network soon after implantation is critical when

engineering an adipose tissue substitute. In Chapter 4, we were successful in rapidly creating a

vascular network by cotransplantation of ASC-embedded modules with a HMEC surface cell

layer. Yet, without the presence of the embedded ASC, HMEC on the surface of the modules

displayed no angiogenic effect. In addition, ASC-embedded modules without HMEC

cotransplantation yielded virtually no fat development. In this chapter we wish to explore the

reciprocal relationship between ASC and EC using a modified in vitro angiogenesis assay and

RT-PCR analysis of several angiogenic and adipogenic genes.

There are numerous in vivo models for studying angiogenesis, which include the chick

CAM assay or injection of Matrigel into mice[128,145]. Yet with these models it is difficult to

purify the EC for biochemical or gene expression analyses. Nehls and Drenckhahn developed a

novel microcarrier-based assay for the study of 3-dimensional cell migration and angiogenesis in

vitro[146]. In their system, EC are cultured as a monolayer on Cytodex beads and then

embedded and cultured in a fibrin gel. This system is well characterized and used extensively in

literature to study EC sprouting and migration. We chose to modify this system, so that instead

of EC covered beads, we used EC covered modules to better mimic the modular constructs that

were implanted in the in vivo studies. Commercially available HUVEC were chosen instead of

HMEC as the EC cell source to minimize the donor-to-donor variability which may affect RT-

PCR results. In Chapter 4, we found that HUVEC-coated modules had a similar angiogenic

response to ASC as did HMEC.

The cross-talk between ASC and EC has been explored in literature, but a complete

understanding of the mechanisms of action are not fully understood. It is believed that a

Page 106: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

92

paracrine relationship exists between ASC and EC. Research over the past decade has identified

some of the pro-angiogenic and anti-angiogenic growth factors expressed by adipose tissues or

adipocytes cultured in vitro. These include VEGF, bFGF, Ang-1,-2, PAI-1, TGF-β and

leptin[89]. We explored the paracrine relationship between HUVEC-coated modules embedded

in the fibrin gel in an in vitro angiogenesis assay and ASC embedded in collagen gel in a hanging

transwell insert. Real time RT-PCR was used to study the effect of coculture on the expression

levels of both adipogenic and angiogenic genes.

5.2.1 Cells 

Adipose-derived stem cells (ASC) were obtained from the stromal vascular fraction of

fresh human abdominal adipose tissue as described in Section 3.2.2. Primary Human Umbilical

Vein Endothelial Cells (HUVEC, Lonza, Walkersville, MD) were cultured in EGM-2 culture

medium (Lonza). Cells were maintained at 37ºC and 5% CO2 in a humidified incubator, medium

was changed every 2-3 days and cells were passaged when ~90% confluent. ASC were used at

passage 3. HUVEC were used between passage 4 and 6.

5.2.2 Optimized In Vitro Angiogenesis Sprouting Assay 

A previously described in vitro microcarrier angiogenesis assay[146,147] was adapted to

study the spouting and migration of HUVEC from the surface of the modules in a fibrin gel, with

and without the presence of ASC in coculture. The experimental set up for the coculture assay is

shown in Figure 5-1. Briefly, a 2mg/mL fibrinogen solution (Sigma, Fibrinogen, Fraction I, type

1-S, from bovine plasma; 85% clottable) was prepared and aprotinin (Sigma, from bovine lung)

was added at 0.15 units/mL. 0.25mL of the fibrinogen solution was added to the bottom of each

well of a 24-well plate containing 0.625U/mL thrombin (Sigma) and pipetted gently to mix and

Page 107: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

93

allowed to form a clot for 20 minutes at 37oC. This was to prepare a bottom layer of fibrin gel so

that modules would not settle to the bottom of the plate. HUVEC-coated collagen modules

(prepared as described in Section 3.2.2) were then gently mixed with the fibrinogen solution (1

package of cut modules/6mL fibrinogen; this is sufficiently dilute to avoid the modules clumping

and forming clusters once in the fibrin gel). 0.5mL of the fibrinogen/module solution was added

on top of the bottom layer of fibrin gel in each well of 24-well plate containing 0.625U/mL of

thrombin (Sigma) and pipetted gently to mix. The plate was placed in a 37oC incubator for 20

minutes to generate the clot. 1mL of EGM-2 (Lonza) was added drop-wise on top of each well

and incubated for at least 30 minutes. The medium was then aspirated and 1-1.25mL of fresh

medium was added to each well. Without the addition of ASC, this set-up was used as the

control.

For the coculture experiments, ASC-embedded collagen gels in hanging transwell inserts

were added to each well containing the HUVEC-coated modules in the fibrin gel. Briefly, ASC

(106cells/mL) were mixed with a 3mg/mL collagen type I solution (PureCol™, Inamed, Fremont,

CA) containing 10X MEM (10.1 g α-Minimum Essential Medium powder (Gibco, Invitrogen,

Paisley, PA), and 2.2g endotoxin free NaHCO3, in 100mL endotoxin free water) and neutralized

to pH ~ 7.4 with 0.8N NaHCO3. 200uL of the collagen-cell suspension was added to the bottom

of cell culture inserts (BD Bioscience, 0.4µm pore size, high density PET membrane) and

allowed to gel at 37oC for 1 hour. 0.3mL of EGM-2 was added to the hanging well insert and

0.7mL of the media was added to the bottom well containing the fibrin gel and cultured in an

incubator at 37oC. The media were changed every 48 hours.

Page 108: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

94

Figure 5-1: Experimental set up of the in vitro angiogenesis assay.

For the coculture experiments, HUVEC-coated modules were embedded in a fibrin gel layer with ASC embedded in a collagen gel in a transwell insert. Control wells consisted of collagen gel in the hanging transwell insert with an empty fibrin gel layer, and HUVEC modules in the fibrin gel layer without ASC in the transwell insert.

5.2.3 Sprouting Counts and Measurements 

The level of sprouting from the HUVEC-covered modules in the coculture set-up was

investigated at 3, 7 and 14 days and compared with HUVEC-covered modules in fibrin gel

without the presence of ASC in the hanging transwell insert as the control. At each time point

investigated, a Zeiss Axiovert 135 (Zeiss, Germany) light microscope equipped with a

CoolSNAP-Pro camera (Media Cybernetics, San Diego CA) using ImpagePro software (Media

Cybernetics) was used at 2.5X objective lens to randomly locate and photograph at least 8

individual modules from both the coculture and control wells. The entire experiment was

repeated 3 times (n=8, N=3). Modules which formed clusters were not used for the analysis due

to difficulty in separating the origin of the sprouts and that the outer surface was blocked by

other modules. Mostly individual modules or modules only slightly merged (ie. 2 modules with

less than ~15% of the surfaces touching) were considered for the analysis. The images were

analysed using the ImageJ (version 1.4.3) software program. A sprout was counted and defined

Page 109: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

95

as a vessel only if it was equal to or greater than 150µm in length. The length of each sprout was

measured as the linear distance from the beginning of the sprout at the module surface to its tip

(Figure 5-2).

Figure 5-2: Image of capillary like-formations growing out from the surface of a HUVEC-coated module in fibrin gel with the presence of ASC in coculture at day 3.

Arrows highlight some counted sprouts, and show the measured linear distance of individual sprouts from the surface of the modules. Scale bar = 250µm.

5.2.4 RNA Isolation 

After photographs were taken for the sprouting measurements, RNA was isolated from

both the HUVEC in the fibrin gel and ASC from the collagen gel in the transwell for RT-PCR

analysis of key angiogenic and adipogenic genes. Experimental set-ups without coculture (e.g,

HUVEC-coated modules in fibrin gel, but without ASC gel in the transwell, and ASC gel in the

transwell but without HUVEC-coated modules in the fibrin gel layer) were used as controls.

RNA was isolated using the RNeasy® Fibrous Tissue Mini Kit (Qiagen). Briefly, gel layers

Page 110: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

96

(either fibrin containing HUVEC or collagen containing ASC) were carefully separated from

individual wells to avoid cross-contamination with the other cell type. A total of 6 gels from

each condition were carefully removed and placed in 1.5mL RNAse free centrifuge tubes. The

samples were centrifuged at 500 x g for 30 seconds to separate the cell and hydrogel from excess

medium. Following the manufacturers instructions, the samples were then homogenized in

300µL buffer RLT (extraction buffer) containing β-mercaptoethanol (Sigma) and treated with a

proteinase K solution at 55oC for 10 minutes to help digest proteins contained in either the fibrin

or collagen gels that may interfere with the RNA purification process. Following centrifugation

at 10,000 x g for 3 minutes the lysate was removed and purified on the RNeasy mini spin

column. The concentration and purity of the RNA was measured using a NanoDrop (ND1000)

Spectrophotometer with a 1µL sample volume. As a measure of purity, the 260/280 ratio was

generally between 1.9 and 2.0, within the desired 1.9 – 2.1 range for RNA.

5.2.5 cDNA Synthesis 

First-strand cDNA was synthesized from 1µg of total RNA using random primers

(Invitrogen; Cat. # 48190-011) and SuperScript™ II Reverse Transcriptase (RT) (Invitrogen;

Cat. # 18064-014). Briefly, from the nanodrop concentration readings, the exact volume of stock

RNA and water required to make up 1µg RNA in 6µL water in 0.2mL reaction tubes was

determined. To each sample, 0.5µL dT primer, 0.5µL random primer and 1.0µL annealing

primer were added and the tubes were heated at 65oC for 5 minutes. After the annealing, 10µL

2x First strand reaction mix and 2µL SuperscriptIII/RNAse out enzyme mix were added to each

tube and the samples were vortex briefly and then centrifuged at 1200 x g for a few seconds.

After 5 minutes of incubation at room temperature, the tubes were put in the PCR thermocycler

Page 111: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

97

(Applied Biosystems 2720), with a cycle set to 37oC (5 min), 50oC (50 min) and then 85oC (5

minutes). cDNA samples were then stored at -20oC until RT-PCR analysis.

5.2.6 Real‐time RT PCR 

The relative expression level of several key adipogenic and angiogenic genes (see tables

for a list of gene specific primers) were used to study the effect of coculture on both ASC and

HUVEC. This was investigated at 72 h, 7 days and 14 days in culture (n=3, N=3). GAPDH

was used as the housekeeping gene for the relative quantification.

The cDNA was diluted in RNAse-free water (Gibco) so that each sample theoretically

contained 10ng of input RNA. The actual amount of cDNA in the sample was dependent on the

efficiency of the cDNA synthesis reactions. Power SYBR® Green Mastermix (Applied

Biosystems, cat# 4367569) was used for the real time analysis. To minimize pipetting errors,

mastermixes were prepared for (i) each of the samples in RNAse-treated water and (ii) the Power

SYBR® Green Mastermix with each of the gene-specific primer sets. Each reaction was

conducted in a well of a 384-well plate with a 10μL reaction volume containing the cDNA

template, 250nM forward primer, 250nM reverse primer and 5µL SYBR® Green Mastermix.

Each gene was analyzed in duplicate for each sample in separate wells.

An Applied Biosystems 7900HT Real-Time PCR system with the SDS 2.3 software

detection program was used for fluorescence detection. The PCR conditions were 95°C for 5

minutes followed by 40 cycles of 30 s at 95°C (denaturation), 30s at 58°C (annealing) and 30s at

72°C (elongation). The fluorescence was measured after each elongation period. A final

extension was conducted for 5 min at 72°C. Following this, melting curve analysis was

conducted to characterize the formed products and confirm that only one amplicon was detected.

For the melting curves, the temperature was increased from 72 °C to 95 °C (held 1 second), with

Page 112: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

98

fluorescence readings every 0.5 °C. Using the software program, the CT (threshold cycle) values

were obtained by analyzing the amplification curves on a logarithmic scale and setting the

threshold within the linear range for all of the genes.

The PFFAFL method[148] for RT-PCR quantification was used to determine the relative

fold-change comparing different samples. The equation for the relative expression ratio is

presented below and is calculated only from the real-time PCR efficiencies and the crossing

point deviation of an unknown sample versus a control. The PCR efficiencies were determined

using the software program LinRegPCR (version 7.2). The program determines a baseline

fluorescence and does a baseline subtraction. Then a Window-of-Linearity is set and PCR

efficiencies per sample are calculated. Unlike the comparative CT method, the PFAFFL model

needs no calibration curve. For HUVEC samples, the treated group is the HUVEC in the

coculture arrangement (ie. HUVEC-coated modules in fibrin gel in the presence of ASC-

embedded collagen gel in the transwell system) while the control is HUVEC-coated modules

cultured without the presence of ASC in the well. This will show the effect of the presence of

ASC on HUVEC gene expression in coculture. For the samples with ASC, the treated group is

ASC in the coculture arrangement (ie. ASC-embedded collagen gel in the transwell with

HUVEC-coated modules in a fibrin gel layer), while the control is ASC-embedded collagen gel

without coculture with HUVEC. This will show the effect of the presence of HUVEC on ASC

gene expression in coculture. Both GAPDH and 18S were used as the housekeeping genes, but

real time results indicated that 18S ribosomal RNA was far more abundant in the samples than

Page 113: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

99

any of the genes investigated. The 18S amplification curves generally crossed the selected

threshold at a very early cycle number, with CT values generally in the range of 5 to 7 cycles.

Since CT values below 10 may be unreliable, GAPDH was used as the housekeeping gene for all

calculations.

Table 2: RT-PCR Primer List

Gene Accession # Description Primers Length (bp)

LPL

M15856 Human lipoprotein lipase • Early marker of adipogenic differentiation. An enzyme secreted by adipocytes into the adjacent capillary endothelium. Hydrolyzes triglycerides into glycerol and free fatty acids. The adipocytes uptake the free fatty acids via a transmembrane protein, where they are processed and stored as triacylglycerols.

F: gtccgtggctacctgtcatt R: tggcacccaactctcataca

94

PPARγ

NM_138712 Human peroxisome proliferator-activated receptor-γ • Early-mid marker of adipogenic differentiation. A nuclear hormone receptor involved in the differentiation of fat. Highly expressed in adipose tissue. Involved in the regulation of adipocyte-specific genes that function in lipid metabolism, such as aP2 and adiponectin. Exists in two isoforms, PPARγ1 and PPARγ2, encoded by a single gene.

F: ttcagaaatgccttgcagtg R: ccaacagcttctccttctcg

88

aP2

J02874 Human fatty acid binding protein • Mid-late marker of adipogenic differentiation. Cytoplasmic protein involved in the intracellular transport and metabolism of fatty acids in adipocytes. Binds fatty acids and transports them within the cell to facilitate triacylglycerol formation and degradation. Upregulated during adipogenic differentiation.

F: tactgggccaggaatttgac R: ggacacccccatctaaggtt

78

GPDH

NM_005276 Human glycerol-3-phosphate dehydrogenase 1 • Late marker of adipogenic differentiation. A cytosolic protein involved in the biosynthesis of fat in adipocytes. Catalyzes the conversion of dihydroxyacetone phosphate (DHAP) (an intermediate in the glycolytic pathway) into glycerol-3-phosphate using NADH as a co-enzyme. Long chain fatty acids form esters with the glycerol-3-phosphate to produce triacylglycerols.

F: gagctgatgcagacaccaaa R: ccacggccactacattcttt

94

Glut4

M20747 Human insulin-responsive glucose transporter • Late marker of adipogenic differentiation. The main glucose transport protein in adipocytes. The presence of insulin activates intracellular signalling cascades that result in the transport of Glut4 transporters from specialized intracellular storage vesicles to the cell membrane, thereby facilitating the transport of glucose into the cell. When insulin levels are reduced, the Glut4 transporters return to the intracellular storage vesicles via endocytosis.

F: agcagctctctggcatcaat R: ctacccctgctgtctcgaag

66

Leptin

NM_000230 Human leptin • Late marker of adipogenic differentiation. A non-glycosylated polypeptide that is secreted by adipocytes as they accumulate lipid. Believed to function in the regulation of body fat. Leptin binding to specialized receptors in the hypothalamus is believed to influence appetite and metabolic rate. Mutations in the gene encoding leptin have been shown to result in obesity in mice.

F: ggctttggccctatcttttc R: accggtgactttctgtttgg

146

Page 114: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

100

Pecam NM_000442

Human platelet/endothelial cell adhesion molecule 1 • EC surface marker Known to be expressed on the surface of endothelial cells, circulating platelets, monocytes, granulocytes and some subsets of T-lymphocytes. PECAM-1 has been proposed as one of the main players in transendothelial migration of neutrophils, monocytes, and natural killer cells in both in vivo and in vitro models

F:aacagtgttgacatgaagagcc R:tgtaaaacagcacgtcatcctt

148

Tie-1 NM_005424

Human Tyrosine kinase with immunoglobulin-like and EGF-like domains 1 • EC surface marker Cell surface protein expressed exclusively in endothelial cells.

F:cacgaccatgacggcgaat R:cggcagcctgatatgcctg

125

PAI-1 M16006

Human Plasminogen Activator Inhibitor 1 A serine protease inhibitor that inhibits fibrinolysis by inactivating urokinase-type and tissue-type plasminogen activator. PAI-1 is also involved in angiogenesis and atherogenesis. PAI-1 is normally secreted by endothelial cells, vascular smooth muscle cells, hepatocytes platelets and adipocytes. Majority of the circulating PAI-1 is contributed to by adipose tissue

F:catcccccatcctacgtgg R:ccccatagggtgagaaaacca

109

bFGF NM_002006

Human basic fibroblast growth factor • Angiogenic growth factor Basic fibroblast growth factor is present in basement membranes and in the subendothelial extracellular matrix of blood vessels

F:atcaaaggagtgtgtgctaacc R:actgcccagttcgtttcagtg

178

PDGFA NM_002607

Human Platelet derived growth factor, subunit A • Angiogenic growth factor Potent angiogenic growth factor involved in the stabilization and maturation of vessels.

F:ccagcgactcctggagataga R:cttctcgggcacatgcttagt

100

VEGF NM_113376

Human Vascular Endothelial growth factor • Angiogenic growth factor Potent angiogenic growth factor known to initiate angiogenesis events.

F:caacatcaccatgcagattatgc R: gctttcgtttttgcccctttc

164

TNFα NM_000594

Human Tumor necrosis factor alpha • Angiogenic growth factor Cytokine involved in systemic inflammation

F:atgagcactgaaagcatgatcc R:gagggctgattagagagaggtc

217

GAPDH NM_002046 Human glyceraldehyde-3-phosphate dehydrogenase • Housekeeping gene.

F: acagtcagccgcatcttctt R: acgaccaaatccgttgactc

94

18S X03205 Human 18S ribosomal RNA • Housekeeping gene.

F: accgcggttctattttgttg R: ccctcttaatcatggcctca

51

5.2.7 Statistical Analysis 

All statistical analyses were performed using the software program STATISTICA Version

5.1 (Statsoft, USA). Student’s t-tests were used to compare the means between two groups

analyzed and considered significant at p<0.05.

Page 115: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

101

5.3 Results 

5.3.1 Effect of ASC on HUVEC Sprouting from Modules in a Fibrin Gel 

The results from the in vitro angiogenesis assay showed a clear relationship between the

presence of ASC in coculture and HUVEC sprouting from the surface of the modules into the

fibrin gel. Figure 5-3 shows representative images of HUVEC sprouting from the surface of the

modules in a fibrin gel, with and without the presence of ASC-embedded gels in a transwell

insert. In the control wells, without ASC, HUVEC could be seen to begin to migrate from the

surface of the modules by day 3 to form short and narrow cord-like structures. These structures

appeared to elongate by day 7 and begin to branch, yet some of the cords appeared to disintegrate

into disorganized groups of individual cells or cell clusters. By day 14, the entire vascular

network that had been previously established had completely degenerated into individual

rounded cells surrounding the modules. In contrast, when an ASC-embedded gel was placed in a

transwell system above the fibrin gel containing the HUVEC-coated modules, a branched

network of HUVEC cords was seen projecting from the surface of the modules as early as day 3.

Branching was thought of as a recapitulation of the sprouting process from the surface of the

module, and was seen extensively in the coculture system. HUVEC cords grew until they

reached another cord and merged. This was seen as a mature and dense network of capillaries by

day 7 with nearby vessels merging to form a vascular network surrounding individual modules.

The diameter of the capillaries at day 7 appeared to be larger than at day 3, suggesting that they

contained lumens surrounded by multiple cells. By day 14, the entire fibrin gel layer contained

an extensive web of vasculature where vessels from one module had reached the vessels of a

nearby module and connected. Again the vessels appeared to be thick with diameters up to

Page 116: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

102

50µm, suggesting that they were multicellular and contained patent lumens (Figure 5-4). No

single cells were seen migrating away from the modules that were not in contact with other cells.

Page 117: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

103

Figure 5-3: Photographs of HUVEC sprouting from modules in a fibrin gel with and without ASC coculture.

With coculture with ASC, HUVEC were seen to sprout from the modules at day 3, the sprouts elongated by day 7 and formed branches and by day 14 a dense vascular network that connected with sprouts from neighbouring was observed. Without ASC coculture, HUVEC were initially seen to form sprouts at day 3 and 7 with minimal branching but these degenerated into a cluster of individual cells surrounding the modules by day 14.

Page 118: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

104

Figure 5-4: Magnified image of HUVEC sprouting from modules with ASC coculture at day 14.

HUVEC sprouts from modules with ASC coculture appeared thick and may be multicellular and contain patent lumens. The arrow points to a sprout that is ~50µm in diameter. Scale bar = 250µm.

Measuring the number and length of HUVEC sprouts growing from the surface of the

modules, coculture with ASC gave a statistically significant improvement in both sprout number

and length as compared to the control case without ASC. As seen in Figure 5-5, there were 15 ±

3 sprouts/module at day 3 which increased to 38 ± 5 sprouts/module for the case with ASC

coculture. Comparing this to the case without ASC, the number of sprouts/module was 7 ± 3 and

13 ± 3 for day 3 and 7 respectfully. The effect of ASC coculture on HUVEC sprouting is clearly

demonstrated by an increase of 25 sprouts/module at day 7 as compared to the control wells

without ASC. Futhermore, by day 14 there were no sprouts to count in the control case and the

branching was too extensive in the coculture case to allow for accurate quantification. As seen

Page 119: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

105

in Figure 5-6 the average length of the sprouts in the coculture case was seen to increase from

227 ± 14µm at day 3 to 491 ± 43µm by day 7, demonstrating that the average sprout length had

almost doubled over this time period. In comparison, without the presence of ASC, the sprout

length did not increase much from 216 ± 22µm at day 3 to 250 ± 19µm by day 7. The average

sprout length in coculture with ASC was on average 240µm longer than the control case without

ASC at day 7.

Figure 5-5: Average number of HUVEC sprouts from the module surface in fibrin gel with and without the presence of ASC in coculture.

A greater number of HUVEC sprouts (>150µm) were observed sprouting from the surface of the modules in the fibrin gel with ASC coculture as compared to control case without ASC (n=8, N=3). Error bars represent ± 1SD. * p < 0.05.

Page 120: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

106

Figure 5-6: Average length of HUVEC sprouts from the module surface in fibrin gel with and without the presence of ASC in coculture.

At day 3 the average length of HUVEC sprouts (>150µm) was nearly identical between the coculture and the control case without ASC. By day 7, the effect of ASC coculture was seen with a near doubling in sprout length as compared to the control without ASC (n=8, N=3). Error bars represent ± 1SD. * p < 0.05.

5.3.2 RT‐PCR Analysis 

Real time RT-PCR analysis of key adipogenic and angiogenic genes was conducted to

help better elucidate the reciprocal relationship between ASC and EC. The transwell system was

used so that the ASC in the collagen gel could be easily separated from the fibrin gel layer

containing HUVEC without chance of cross-contamination between the cell types.

The data presented here in Figure 5-7, Figure 5-8 and Figure 5-9 represents the relative

fold change in mRNA expression of either 1) ASC in coculture vs. ASC alone, or 2) HUVEC in

coculture vs. HUVEC alone. Due to the variable nature of the obtained results, each

experimental value from each of the three trials is plotted. The variable nature of the results may

be due, in part, to donor cell variability. Two of the experiments were conducted with ASC

derived from the same patient, while the other was from another donor. The data points derived

from the same patient are visualized on the graphs as a solid colour point, while the other data

Page 121: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

107

point is not filled with colour. A significant fold change in mRNA expression level is considered

to be anything greater than a 1.5 or less than 0.5.

From the ASC results (Figure 5-7), it appears that these cells were not being induced to

differentiate by the HUVEC under the experimental conditions examined here. If so we would

have expected at least some of these adipogenesis markers to be clearly expressed more. While

there was some evidence that LPL (an early marker of adipogenesis) was expressed more than

without HUVEC coculture at day 3, this is back down to similar levels by day 7 and 14.

Interestingly, both PPARγ and aP2 appear to be expressed less at day 3 and then return to

unchanged levels by day 7 and 14. The only clear example of a change in mRNA expression for

all 3 experimental trials is Glut4 which demonstrated fold-changes greater than 2 at day 3, but

then again it is back to unchanged levels by day 7 and 14.

Page 122: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

108

ASC (Adipogenic Genes)

LeptinGlut 4GPDHaP2PPARLPL0

0.5

1

1.5

2

2.5

3

3.5

4

Rela

tive

Fold

Cha

nge

in m

RNA

Day 3Day 7Day 14

Figure 5-7: Relative fold change in mRNA expression of adipogenic genes of ASC in coculture with HUVEC, vs. ASC cultured alone.

HUVEC coculture does not appear to induce an adipogenic phenotype of the ASC in the in vitro conditions investigated here. Only aP2 expression appears to be decreased and Glut4 expression increased for all 3 trials at day 3. Solid colour data points represent 2 trials with the same donor ASC. The unfilled data point represents ASC from a different donor.

Exploring the angiogenic genes investigated (Figure 5-8), there was a significant increase

in PAI-1 expression at day 3, with unchanged levels at day 7 and 14. Interestingly, HUVEC did

not seem to change the expression levels of bFGF or PDGF in ASC at any of the time points

investigated. VEGF expression was clearly increased at day 3, with fold change values greater

than 3.5, but returned to unchanged levels by day 7 and 14. ASC from two of the trials (same

donor, solid colour data points) appeared to be have increased expression in TNFα with fold

Page 123: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

109

changes greater than 1.5 at days 3 and 7, while fold change in ASC from the other donor

(outlined data points) were unchanged.

ASC (Angiogenic Genes)

PAI 1 bFGF PDGF VEGF TNFa0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

5.5

Rela

tive

Fold

Cha

nge

in m

RNA

Day 3Day 7Day 14

Figure 5-8: Relative fold change in mRNA expression of angiogenic genes of ASC in coculture with HUVEC, vs. ASC cultured alone.

Coculture with HUVEC increased the expression of PAI-1, VEGF and possibly TNFα (one donor) at early timepoints. These factors may be involved in ASC-EC paracrine signalling. Expression of all other genes remained unchanged. Solid colour data points represent 2 trials with the same donor ASC. The unfilled data points represents ASC from a different donor

Examining the results of the HUVEC cells and the impact that ASC coculture had on the

relative gene expression (Figure 5-9), we can see that ASC had little impact on the EC markers

PECAM and Tie-1, with only PECAM at day 7 showing fold-changes greater than 1.5 for all

three experimental trials. Surprisingly, PAI-1, bFGF and PDGF all showed little change in

Page 124: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

110

mRNA expression with possibly the exception of bFGF which had quite variable results. It

appeared that VEGF at day 3 and 14 may be upregulated in HUVEC in the coculture system.

HUVEC

VEGFPDGFbFGFPAI 1Tie1PECAM0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

Rela

tive

Fold

Cha

nge

in m

RNA

Day 3Day 7Day 14

Figure 5-9: Relative fold change in mRNA expression of HUVEC in coculture with ASC, vs. HUVEC cultured alone.

Coculture with ASC increased the expression of bFGF (one donor) at day 3 and VEGF at day 3 and 7 (one donor). Expression of all other genes was variable and unchanged. Solid colour data points represent 2 trials with the same donor ASC. The unfilled data point represents ASC from a different donor.

Page 125: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

111

5.4 Discussion 

The results from Chapter 4 of this thesis showed a clear relationship between ASC and

EC in vivo. Only with ASC cotransplantation inside the modules, were the HMEC able to detach

from the surface of the modules, organize into vessels, anastomose with the host vasculature and

remain viable for up to 90 days. It appeared that the ASC promoted EC survival and had an

angiogenic effect on EC in vivo. The objective of this work was to conduct a preliminary study

to help elucidate the crosstalk between these two cell types in vitro.

The results from the in vitro angiogenesis assay recapitulated what was seen in vivo. The

presence of ASC in the transwell led to extensive sprouts and tube formation from the HUVEC

on the surface of the modules, leading to a branched and interconnected network inside the fibrin

gel layer by day 14. In contrast, without the presence of ASC, HUVEC were found to initially

form some sprouts at early time points, but by day 14 complete degeneration of the vascular

network was observed and only individual cells were seen surrounding the modules.

The in vitro sprouting assay as developed by Nehls and Drenckhahn[146], typically

involves culturing EC on Cytodex beads and then embedding these beads in a fibrin gel. They

showed that this technique works well for bovine aortic EC (BAEC), which were shown to bud

and form capillary-like structures in response to angiogenic growth factors such as bFGF and

VEGF. However, there may be phenotypic differences in the type of EC used as their sprouting

assay did not show sprouting from HUVEC. One group has modified this protocol to include

skin fibroblasts cultured on top of the gels containing the HUVEC covered beads[149]. They

found that only in the presence of these fibroblasts were the HUVEC able to sprout, divide,

migrate, form lumens and connect with neighbouring vessels, similar to what is observed in

angiogenesis in vivo. ASC share several phenotypic similarities to fibroblasts so that it is not

Page 126: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

112

surprising that we see with ASC what was seen with fibroblasts. In a recent article, coculture

of ASC and HUVEC was studied in monolayer coculture on tissue culture plastic. They found

that while both fibroblasts and smooth muscle cells promoted HUVEC tube formation in

coculture, ASC promoted vascular network formation much more strongly based on total tube

length and network branching[91]. It appeared that soluble factors secreted by a supporting cell

are needed to encourage HUVEC sprouting and maturation into a stable vascular structure in

vitro and that the paracrine relationship between ASC and EC has a stronger angiogenic effect.

But the question remains what is this paracrine relationship? Are the paracrine effects the only

contribution to the observed angiogenic effect on EC both in vivo and in the in vitro angiogenesis

assay?

The paracrine effects between adipogenesis and angiogenesis have been studied in the

literature, typically by blockade of an angiogenic growth factor or knock-out of a specific

angiogenic gene but there is no clear consensus in the literature as to what is really happening.

VEGF and hepatocyte growth factor secreted by ASC, and PDGF of endothelial origin, have

been shown to be essential for ASC-EC vascular network formation[91]. Knock-out of PPARγ

(master regulator of adipogenesis) has been shown to not only abrogate fat development in vivo,

but angiogenesis as well[150]. The RT-PCR results obtained here do not support the notion that

HUVEC have an effect on the mRNA expression levels in the ASC. This may not be surprising

as ASC differentiation in vitro is conducted in serum-free medium containing a defined cocktail

of adipogenic factors. Perhaps the EGM-2 media used in the coculture experiments contained

growth factors, such as bFGF, at concentrations that promote a proliferative phenotype of the

ASC. This was observed qualitatively, as ASC cultured on TCPS in EGM-2 appeared to grow

much more rapidly and reach confluence sooner than ASC cultured in the standard DMEM/F12-

Page 127: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

113

Ham’s media. For the angiogenic genes, expression of PAI-1, TNFα, and VEGF all appeared to

be increased at early timepoints in the ASC in the coculture system. These three growth factors

are hypoxia-inducible angiogenic factors that are known to be increased with obesity[151,152].

Interestingly expression of these three growth factors were reported to increased at early

timepoints in an in vivo model of adipose tissue development[150]. Yet without further

evidence, it is impossible to say if the increase in mRNA expression of these three growth factors

at early time points is responsible for downstream angiogenic events.

The gene expression from HUVEC, revealed no clear impact of ASC coculture besides

increase in VEGF expression at day 3 and 14. Increased expression of VEGF is not surprising as

it is a potent angiogenic growth factor responsible for the initiation of the angiogenic cascade

and the most studied growth factor in the paracrine effect between adipogenesis and

angiogenesis. It appeared that the RT-PCR data does not agree with the observed increase in

HUVEC sprout number and length with ASC coculture. Perhaps, other genes than those

investigated are involved in the observed effects including genes involved in cell apoptosis (e.g.

caspase-3, p53).

In the in vitro angiogenesis assay we studied only the paracrine effects of ASC-EC

interaction since there was no contact between the two cell types. Perhaps this is not the only

mechanism behind the observed ASC-EC interaction. Merfeld-Clauss et al. have shown that

ASC are localized in the peri-endothleial layer in native fat tissue, and can acquire properties of

pericytes both in vitro and in vivo[87,129]. In their in vitro studies they showed that vascular

network density was 60% lower for EC in direct contact with human dermal fibroblasts than that

observed for EC in direct contact with ASC in the very same well. They went on to show that

ASC were localized in proximity to the EC cords and that the areas distant from EC cords were

Page 128: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

114

clearly depleted with ASC[91]. The ASC that were adjacent to EC showed an increase in α-

smooth muscle actin expression and organization into fibres. Perhaps, the EC were

differentiating the ASC into pericyte-like cells that are able to stablize the growing vasculature.

While these are only recently published observations, it appears that both paracrine

communication and physical contact between ASC and EC may be necessary for the sequence of

events leading to the initiation, growth and stabilization of vascular structures both in vitro and

likely in native adipose tissue in vivo. We observed an increase in both HUVEC sprout number

and length with ASC cocultre, even without direct ASC-EC contact. Perhaps, the paracrine

effects are sufficient to establish a vascular network while pericytes further strengthen the

developing vasculature into stable vessels that can support blood flow.

5.5 Conclusions 

Using an in vitro angiogenesis assay we have shown that the presence of ASC in

coculture promotes an angiogenic phenotype (sprouting and migration) of HUVEC. HUVEC in

the presence of ASC, were seen to sprout from the surface of the modules by day 3, elongate and

branch by day 7 and form a dense network of vessels by day 14 that merged with sprouts from

nearby modules. In contrast, HUVEC without ASC coculture initially formed sprouts, but these

degenerated into a mass of invidual cells surrounding the modules by day 14. RT-PCR analysis

of key adipogenic and angiogenic genes was variable, possibly due to different ASC donors. No

clear difference in mRNA expression of adipogenic genes was observed for ASC in HUVEC

coculture. Increased expression of PAI-1, VEGF and TNFα was observed in ASC with coculture

with HUVEC at day 3, and these may be important players in the paracrine signaling between

ASC and EC. The gene expression from HUVEC revealed no clear impact of ASC coculture

besides upregulation of VEGF at day 3 and 14. Recent reports in the literature highlight that not

Page 129: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

115

only paracrine signaling, but direct ASC-EC contact may be important in creating a vascular

network as ASC possess characteristics of pericytes.

5.6 Acknowledgements 

The authors acknowledge the financial support by NSERC. Special thanks to Lindsay

Fitzpatrick for her help with RT-PCR.

Page 130: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

116

6 Synopsis 

Modular tissue engineering represents a shift in the traditional cell + scaffold paradigm.

Producing small sub-mm sized hydrogel modules embedded with functional cells and coated

with a layer of EC is a means to produce scalable, vascularized tissue in vivo. For adipose tissue

engineering, the modules are embedded with ASC and surface coated with HMEC. We imagine

thousands of these modules implanted at the site of a soft tissue defect to restore normal

cosmesis. Once the modules are implanted, the transplanted HMEC are expected to quickly

form a vascular network that anastomoses with the host, while the ASC differentiate into mature

adipocytes resulting in a fat pad. This project has addressed the two main reasons where other

approaches to adipose tissue engineering have failed, mainly; 1) insufficient mechanical strength

of the cellular support structure and 2) insufficient vascularization upon implantation to support

transplanted cells. We explored the use of a semi-synthetic collagen/poloxamine hydrogel to use

as a biomaterial for adipose tissue engineering by testing its ability to support ASC viability and

differentiation capacity and EC attachment to the surface. Next we investigated the ability of the

modular approach to produce vascularized adipose tissue in vivo that persisted for the long-term

(3 months). Then to better understand the angiogenic remodeling of the transplanted HMEC in

vivo, we investigated the paracrine relationship between ASC and EC in an in vitro model of

angiogenesis with RT-PCR analysis of key angiogenic and adipogenic genes. This chapter

highlights some of the lessons learned and gives suggestions of future experiments and

strategies.

Page 131: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

117

6.1 Biomaterials for Adipose Tissue Engineering 

To successfully create a tissue engineered construct, a support structure is needed to serve

as a site for cellular attachment and guidance in three-dimensions. Not only must this support

structure possess the needed biological properties to promote the desired function of the

transplanted cell, but in adipose tissue engineering it must also demonstrate favorable

mechanical properties as well. Rigid or stiff materials (ie. PLGA) that are beneficial for load

bearing applications are not suitable for soft tissues where the esthetic shape and feel is most

important. Degradation of the scaffold material is also important. If the scaffold does not

degrade quickly enough, host fibrous tissue may impair differentiation of transplanted ASC and

vascular ingrowth. On the other hand, rapid degradation of the scaffold may result in the loss of

the desired three-dimensional shape before the maturation of the fat pad has occurred. An

appropriate balance between scaffold degradation and adipose tissue accumulation must be

achieved.

Hydrogels are considered optimal for adipose tissue engineering because they mimic the

feel of native adipose tissue most closely. Often they can be injected at the site of the tissue

defect in a minimally invasive procedure. In the module approach, while the polymer was not

injected and polymerized in situ, the modules were small enough to be injected through an 18-

gauge needle which is advantageous in a clinical setting. Yet, synthetic polymers are

disadvantageous in that they typically are not cell adhesive and must be modified to contain

bioactive moeities. The collagen/poloxamine hydrogel, for example, was unsuccessful as a

biomaterial for adipose tissue engineering (Chapter 3). Simple mixing of the collagen and

poloxamine before photopolymerization resulted in a network of the two components where

entropy-driven phase separation resulted in regions in the hydrogel devoid of collagen that

Page 132: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

118

spanned distances greater than 100µm. ASC that were in these regions were not viable and did

not differentiate into adipocytes. We originally hypothesized that the 2-phase network of

collagen and poloxamine was advantageous for ASC differentiation. While ASC are anchorage-

dependent cells, adipocytes may prefer a less cell adhesive environment as they are known to

detach from TCPS during lipogenesis in in vitro culture. We thought that the ASC would attach

to regions in the hydrogel rich in collagen and during adipogenesis cell-secreted collagenase

would degrade the collagen so that mature adipocytes would be in the less cell adhesive

poloxamine phase. Yet, because of extent of phase separation between the collagen and

poloxamine components, many cells were only in the poloxamine phase and died. It appears that

at least some initial attachment to the matrix is needed to support ASC viability before

differentiation commences. We could explore the nature of ASC differentiation in a non-

adhesive substrate by using collagen and poloxamine in a slightly different arrangement. ASC

could be embedded in small amounts of collagen gel (ie. collagen microparticles) which are

subsequently photopolymerized within a poloxamine matrix. The ASC would initially be in

collagen to enable attachment and proliferation, yet during lipogenesis the cell secreted

collagenase would dissolve the collagen support and the resulting adipocytes would be in the

non-adhesive poloxamine. Oil Red O staining and RT-PCR analysis of adipogenic genes could

be used to determine the effect of the substrate on differentiation as compared to a collagen

control.

For a synthetic hydrogel to work as a biomaterial for adipose tissue engineering it must

be tailored to support both ASC attachment and proliferation onto the matrix and subsequent

differentiation into adipocytes as well. Poloxamine could be chemically modified to contain cell

adhesive molecules such as RGD or YIGSR that would enable ASC attachment and spreading

Page 133: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

119

within the matrix as well as EC surface attachment. In addition, a degradable version of

poloxamine must be investigated as well. Optimized degradation of the matrix would occur if

degradation was controlled by the differentiation of the ASC. This would ensure that the matrix

does not degrade too quickly before maturation of the fat pad has been achieved. The

collagenase-sensitive peptide sequence Leucine-Glycine-Proline-Alanine (LGPA) as investigated

by Jennifer West and colleagues[63] could be incorporated into the poloxamine to allow cell-

mediated degradation of the matrix during adipogenesis. Poloxamine, incorporating both

YIGSR and LGPA, appears to be a better approach to enabling ASC attachment, proliferation

and differentiation as well as degradation of biomaterial and may have future applications in

adipose tissue engineering.

6.2 Revascularization Strategies 

Rapid vascularization of tissue engineered constructs upon implantation is critical to

support transplanted cells and integration of the device with the host. The lack of vascularization

is the main reason why both autologous fat transfers and previous approaches to tissue

engineered adipose constructs have been unsuccessful in repairing soft tissue deficits. We

demonstrated that the modular approach to adipose tissue engineering is successful in rapidly

creating a vascular network and promoting long-term fat development (3 months) in a construct

of size greater than the diffusional limitation of oxygen and nutrient transfer. Our approach has

the advantage that both ASC and HMEC are derived from the same patient, meaning that the

adipose tissue engineered construct can be an autologous transplant and hence may avoid

immunogenic rejection.

Other reports have demonstrated that EC transplantation as a revascularization strategy

can only be successful with transfection of an anti-apoptotic gene or with the presence of a

Page 134: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

120

supporting cell[119,122]. We found that without ASC cotransplantation, HMEC on the surface

of the modules did not result in a new vasculature and died over 14 days in vivo, even in the

immunodeficient SCID mouse used as the animal model here. Yet, with ASC embedded inside

the modules, the HMEC on the surface quickly detached and formed primitive vessel-like

structures that contained erythrocytes as early as day 3, suggesting inosculation with the host.

As time progressed, the number of vessels decreased but the size of the vessels increased

indicating vessel maturation and connection with the host was proved by Microfil perfusion at

day 21. It appeared that the ASC had an angiogenic effect on the transplanted HMEC and

promoted HMEC viability. This was not unique to HMEC as implants with HUVEC revealed a

similar angiogenic response.

The angiogeneic effects may be explained by the paracrine relationship between ASC

and EC. As discussed in Chapter 5, ASC are known to produce EC-specific mitogens and

angiogenic factors, while EC produce factors that stimulate ASC proliferation and

differentiation[89]. Yet, recent reports have suggested that ASC are localized in the peri-

endothelial layer of native adipose tissue and may be able to aquire properties of pericytes both

in vitro and in vivo following harvesting and expansion[87,129]. The hypothesis here is that cell

contact between the ASC ‘pericytes’ and the EC may help to stabilize vascular network

formation. While EC vessels may initially form, without this mural layer (ie. pericytes or

smooth muscle cells) these small vessels may degrade followed by EC death, which is what was

observed in the case without ASC cotransplantation.

It would be interesting to study and track the fate of the ASC in vivo to see if

ASC/pericyte vessel stabilization is responsible for the formation of the vascular network. ASC

could be harvested from a green fluorescent protein (GFP) expressing mouse so that we could

Page 135: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

121

see if the ASC were migrating from the inside of the modules and then localizing at the vessel

walls of the HMEC-derived vessels.

While there is a lack of known ASC specific stem cell markers that could be used to track

the fate of these cells in vivo, there is some evidence that subpopulations in the stromal vascular

fraction of adipose tissues may give rise to different cell types. CD34- populations have been

shown to express more pericyte markers and may be the subpopulation of ASC that differentiate

into pericytes[87,88]. We could use flow cytometry to separate different subpopulations of ASC

and use these cells embedded into the modules. We would then compare the effect of CD34-

enriched populations of ASC on vessel formation in vivo. If ASC-EC cell contact proves

important in the vascular remodeling process, we could design modules that contain embedded

ASC with a mixture of HMEC and ASC attached on the surface. Perhaps this would accelerate

the remodeling of the HMEC-derived vascular network because supporting ASC would not have

to migrate from inside the collagen gel to the module surface.

While the understanding of why ASC promote and support HMEC vessel formation is

unclear, the formation of a rapid vascular network is needed to support transplanted cells. Only

in the case of cotransplantation did we see fat accumulation inside the modules. ASC-only

modules without HMEC on the surface showed little to no visible signs of fat accumulation by

histological analysis. This is likely the result of a lack of sufficient blood vessel network to

nourish the implant, but may also be contributed to by the paracrine effects of EC/ASC

interaction. With the lack of an appropriate stem cell marker for ASC, we cannot confirm that

the fat accumulation is due to transplanted ASC and not host cells, but the location of the fat

deposits seen inside individual modules is compelling. In the case of cotransplantation, fat

accumulation was seen to begin at day 30 and increase until day 90, yet the volume of the

Page 136: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

122

implants decreased over time. This is not surprising as collagen gel was used as the module

material and degrades too quickly to be used by itself as a biomaterial for adipose tissue

engineering. In an in vivo experiment, it would be interesting to strengthen the collagen gels,

eg., by crosslinking the gels by transglutaminase[153], to see if stiffer collagen modules when

implanted would degrade more slowly so that maturation of the fat pad happens before the

support structure has degraded to promote a long-term volume stable mass of adipose tissue.

6.3 Understanding the relationship between ASC and EC 

While the in vivo experiments showed qualitatively that ASC had an angiogenic effect on

transplanted EC, we further explored the relationship between ASC and EC in vitro to help

elucidate the paracrine relationship between the two cell types. Adapting an in vitro

angiogenesis model as developed by Nehls and Drenckhahn[146], we embedded HUVEC coated

modules inside a fibrin gel with ASC-embedded in a collagen gel separated in a transwell so that

no ASC-EC direct contact occurred. The primary reason for this was to ensure easy separation

of the two cell types for RT-PCR analysis. In accordance with the in vivo results, HUVEC were

seen to sprout and form a dense vascular network over 14 days in the fibrin gel with the presence

of ASC. In the wells without the ASC, HUVEC initially formed some sprouts but these began to

degenerate by day 7 and by day 14 only an unorganized cluster of individual cells was seen

surrounding the modules. Without any direct ASC-EC contact this is an example of paracrine

signaling between the two cell types that helped establish and mature the HUVEC-derived

vascular network.

As discussed in the previous section, it would be interesting to explore the role of ASC as

pericytes to support the growing vascular network. As evidenced here, ASC as pericytes (ie.

with direct contact) were not needed to successfully form a dense sprouting network of HUVEC

Page 137: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

123

in fibrin gels, yet ASC as pericytes may (further) aid the vascular network formation in vivo. As

a next experiment, we would use ASC-embedded modules coated with HUVEC, or a mixture of

ASC and HUVEC on the module surface in the in vitro angiogenesis assay to explore the effect

of ASC-EC contact on vascular network formation. We would label the ASC with a fluorescent

dye (ie. DiI) and the EC could be tracked with CD31 antibody staining. Using confocal

microscopy we could locate the presence of ASC surrounding the HUVEC sprouts to see if both

cell types were colocalized suggesting that ASC is supporting the developing vasculature.

Others have also found that α-smooth muscle actin (αSMA) expression is upregulated in EC-

ASC cocultures[91]. αSMA antibody staining could be used to see if it is upregulated in ASC

with close proximity to the HUVEC and its organization into fibres.

The sprouting results showed the clear impact that the ASC had on the vascular network

formation of HUVEC from modules, so it was expected that RT-PCR analysis of key adipogenic

and angiogenic genes from these experiments would further confirm these observations. The

coculture with HUVEC resulted in little change in mRNA expression of early, mid and late

markers of adipogenesis in the ASC. Perhaps, the medium used in this experiment (EGM-2)

masked any possible impact because it contained numerous growth factors that promote a more

proliferative phenotype of ASC. Growth arrest (ie. serum free medium) is normally a

prerequisite for ASC differentiation in vitro. For the angiogenic factors investigated, VEGF,

PAI-1 and TNFα expression were increased in ASC at early timepoints with HUVEC coculture.

These three factors are likely players in the paracrine relationship between ASC-EC but without

further investigation (e.g. inhibition of adipogenesis or angiogenesis with a blocking antibody, or

gene transfection) this cannot be concluded here. The data tended to be quite variable, which

may be the result of ASC donor variability. Two of the experiments were conducted with ASC

Page 138: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

124

from the same donor and the results of those two experiments appeared to be less variable for

many of the genes investigated.

For HUVEC, it appeared that ASC coculture resulted in increased expression of VEGF at

early time points which is not surprising as VEGF is a potent initiator in the angiogenic cascade.

Again the results tended to be variable making it difficult to draw conclusions. Perhaps, instead

of looking at a small subset of genes a gene array should be used to look at multiple genes at

once. The TaqMan “Human Angiogenesis Array” contains 94 angiogenic targets, including

angiogenic growth factors, inhibitors, markers and targets and may provide a better insight into

the angiogenic remodeling of HUVEC by ASC. It would be interesting to also test genes

involved in cell apoptosis since the ASC were shown to promote HMEC viability in vivo.

6.4 Future Directions 

The future of adipose tissue engineering is a bright one. There are two current challenges

that need to be addressed before an adipose tissue engineered construct can move from the

bench-top to the clinic:

1) A suitable biomaterial that possesses the necessary cell adhesive properties to allow

ASC attachment, proliferation and differentiation as well as the required mechanical

properties that give an appropriate ‘feel’ and degradation in the body and;

2) A successful revascularization strategy to nourish transplanted cells to encourage ASC

survival and differentiation into a mature fat pad that will retain its volume and shape.

In future work, biomaterial design will likely focus on cell-ECM interactions to provide

biological cues that will guide and direct ASC growth and differentiation. The interactions with

ASC and either synthetic or natural polymers, with and without soluble factors, could be used to

Page 139: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

125

create a biological niche that will direct cell fate. Cell shape, for example, has been shown to

drive MSC commitment. MSC cultured on fibronectin islands of restricted size (1,024µm2) were

shown to remain rounded and differentiated into adipocytes, while those cultured on larger

islands (10,000µm2) were able to spread and differentiated into osteoblasts[140]. Restricting the

cells from spreading abrogates the ability to form geometrically separated adhesions, where

when the cells are unrestricted they can form many focal adhesions that act on tensioning of the

cytoskeleton. For a hydrogel, the effect of tensioning of the cytoskeleton is not controlled by the

available surface area for attachment, since the cells are freely embedded or coated on the

surface, but rather the substrate elasticity. MSC cultured on soft substrates (Elastic modulus

(E)~0.1-1kPa) that closely mimic the mechanical properties of soft tissue were shown to highly

express neurogenic markers, while stiffer substrates with E~8-17kPa and E~25-40kPa were

shown to preferentially express myogenic and osteogenic markers respectively[154]. From this

study it appears that the elasticity of the microenvironment that closely matches that of the native

tissue helps drive differentiation into that particular lineage, so therefore a soft hydrogel is likely

optimal for ASC differentiation. Yet, substrate elasticity is not the only factor that can control

stem cell fate. While increases in gel stiffness from 0.7kPa to 80kPa increased MSC spreading

areas from 5,000 to 45,000µm2, those coated with laminin had only half of the spreading area

compared to those coated with fibronectin of the same stiffness[155]. This study shows that

incorporation of bioactive ligands into the biomaterial is another approach to direct stem cell

fate. In a recent publication, differentiation of MSC embedded in a PEG hydrogel was

controlled by incorporation of small-molecule functional groups. MSC encapsulated in a PEG

hydrogel containing charged phosphate groups were shown to undergo osteogenesis while those

cultured in hydrogels containing t-butyl groups differentiated into adipocytes[156]. These

Page 140: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

126

studies highlight that both mechanical properties and the incorporation of bioactive

ligands/molecules are key design considerations when developing a biomaterial for adipose

tissue engineering. To help with biomaterial design, improved characterization of ASC and

understanding of the immunophenotype of the different populations of adipose progenitor cells is

needed. Furthermore, understanding the impact of in vitro cell conditioning of ASC prior to

implantation (ie. predifferentiation) may also affect the in vivo outcome.

Cotransplantation of ASC and EC in the modular approach appeared to be a successful

revascularization strategy in the work presented here. Yet further understanding of the ASC-EC

interaction on the molecular level is needed to fully exploit its potential. The capability of ASC

to not only differentiate into adipocytes but also endothelial cells and pericytes is not well

understood but is likely key to understanding ASC-EC vascular network formation in vivo. We

must look to the biology of the complex interplay between adipogenesis and angiogenesis for

answers. In addition to better understanding of the cells, is to better understand the implant

model to test the outcome of an engineered adipose construct. The majority of the adipose tissue

engineering studies to date have been subcutaneous implantation into rodent models. The next

step is to investigate other implant sites in larger animal models to study the remodeling of an

adipose tissue engineering construct of clinical relevance. All in all, to move adipose tissue

engineered constructs into clinical practice standardization of cell sourcing, biomaterial design,

and implant procedures is needed.

Looking at the bigger picture, an ASC-EC revascularization strategy may have widespread

application not only for adipose tissue engineering, but tissue engineering as a whole. Current

interest in the field of islet transplantation, for example, is the use of adipose tissue and ASC to

create a vascular network that will support islet engraftment. A recent publication demonstrated

Page 141: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

127

the use of ASC combined with minced adipose tissue to create vascular-rich beds that were

suitable to support islet transplantation subcutaneously in diabetic mice. They were able to

correct blood glucose level within a week after islet transplantation and maintain normoglycemia

for over 8 weeks[157]. We can imagine combining ASC-EC modules with modules containing

islets where once implanted the EC rapidly create a functional vascular network that will support

the viability of the transplanted islets as a treatment for diabetes.

Page 142: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

128

7 Conclusions 

The overall objective of this project was to build a modular approach for adipose tissue

engineering to create vascularized adipose tissue in vivo that persisted long-term (3 months). In

addition we wanted to better understand the mechanism behind the vascularization seen with

transplantation of ASC in EC covered modules. We explored and tested three different

hypotheses:

1. Fabricate and optimize the modular platform for adipose tissue engineering to promote

embedded ASC viability, differentiation and attachment of surface coated endothelial cells

(EC).

Hypothesis: The semi-synthetic collagen/poloxamine hydrogel used as the module material

promotes high viability of embedded ASC and allows differentiation of these cells into mature

adipocytes, while enabling good adhesion and coverage of EC on the module surface.

Rejected: Entropic phase separation between collagen and poloxamine resulted in a 2-phase

biomaterial. ASC were not able to attach, proliferate and differentiate into regions of

the matrix rich in poloxamine. EC were unable to form a confluent layer on the

surface of the biomaterial.

2. Evaluate the regenerative potential of the modular construct to produce vascularized adipose

tissue in vivo.

Hypothesis: The endothelialized perfusion channels created by packing many modules into a

larger construct provide an immature capillary network that remodels and anastomoses with the

Page 143: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

129

host vasculature upon implantation to promote transplanted ASC viability and facilitates a long-

term (3 months) volume-persistent culture of adipose tissue in vivo.

Accepted: Cotransplantation of ASC and HMEC in the modular approach resulted in rapid

vascular network formation that appeared to connect with the host vasculature. An

accumulation of adipose tissue was created that persisted for up to 3 months in vivo.

3. Investigate ASC/EC interaction in an in vitro sprouting assay.

Hypothesis: Coculture of ASC and HUVEC (no direct cell-cell contact) promotes an angiogenic

phenotype (i.e. migration and sprouting) of surface covered HUVEC in a fibrin gel.

Accepted: Coculture of ASC and HUVEC in an in vitro sprouting assay resulted in a dense

vascular network formation of HUVEC surrounding the modules within a fibrin gel.

HUVEC without ASC coculture showed initial sprouting and migration but this

network disintegrated over 14 days in culture. RT-PCR analysis demonstrated that

VEGF, PAI-1 and TNFα expression increased in coculture and may be involved in

paracrine signaling between ASC and EC.

The modular approach to tissue engineering was successfully applied to create

vascularized adipose tissue in vivo that persisted for at least 3 months. The success of the EC

transplantation revascularization strategy is attributed to, at least in part, to paracrine interactions

between ASC and EC. This work has potential in creating a clinically relevant adipose tissue

substitute. ASC-EC revascularization strategy may have widespread use in tissue engineering

applications.

Page 144: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

130

8  Reference List 

Reference List

1. Ersek RA: Transplantation of purified autologous fat: a 3-year follow-up is disappointing. Plast Reconstr Surg 1991, 87: 219-227.

2. Niechajev I, Sevcuk O: Long-term results of fat transplantation: clinical and histologic studies. Plast Reconstr Surg 1994, 94: 496-506.

3. Awwad HK, el Naggar M, Mocktar N, Barsoum M: Intercapillary distance measurement as an indicator of hypoxia in carcinoma of the cervix uteri. Int J Radiat Oncol Biol Phys 1986, 12: 1329-1333.

4. Patrick CW: Breast tissue engineering. Annu Rev Biomed Eng 2004, 6: 109-130.

5. Lanza RP, Langer RS, Vacanti J: Principles of Tissue Engineeing, 2nd edn. San Diego, CA: Academic Press; 2000.

6. Gregoire FM, Smas CM, Sul HS: Understanding adipocyte differentiation. Physiol Rev 1998, 78: 783-809.

7. Petruschke T, Hauner H: Tumor necrosis factor-alpha prevents the differentiation of human adipocyte precursor cells and causes delipidation of newly developed fat cells. J Clin Endocrinol Metab 1993, 76: 742-747.

8. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM: Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 1995, 95: 2409-2415.

9. Hirokawa J, Sakaue S, Tagami S, Kawakami Y, Sakai M, Nishi S et al.: Identification of macrophage migration inhibitory factor in adipose tissue and its induction by tumor necrosis factor-alpha. Biochem Biophys Res Commun 1997, 235: 94-98.

10. Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-Vague I: Production of plasminogen activator inhibitor 1 by human adipose tissue: possible link between visceral fat accumulation and vascular disease. Diabetes 1997, 46: 860-867.

11. Smas CM, Sul HS: Control of adipocyte differentiation. Biochem J 1995, 309 ( Pt 3): 697-710.

12. Niemela SM, Miettinen S, Konttinen Y, Waris T, Kellomaki M, Ashammakhi NA et al.: Fat tissue: views on reconstruction and exploitation. J Craniofac Surg 2007, 18: 325-335.

Page 145: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

131

13. Avram MM, Avram AS, James WD: Subcutaneous fat in normal and diseased states 3. Adipogenesis: from stem cell to fat cell. J Am Acad Dermatol 2007, 56: 472-492.

14. Rosen ED, MacDougald OA: Adipocyte differentiation from the inside out. Nat Rev Mol Cell Biol 2006, 7: 885-896.

15. Feve B: Adipogenesis: cellular and molecular aspects. Best Pract Res Clin Endocrinol Metab 2005, 19: 483-499.

16. Smith PJ, Wise LS, Berkowitz R, Wan C, Rubin CS: Insulin-like growth factor-I is an essential regulator of the differentiation of 3T3-L1 adipocytes. J Biol Chem 1988, 263: 9402-9408.

17. Girard J, Perdereau D, Foufelle F, Prip-Buus C, Ferre P: Regulation of lipogenic enzyme gene expression by nutrients and hormones. FASEB J 1994, 8: 36-42.

18. Marques BG, Hausman DB, Martin RJ: Association of fat cell size and paracrine growth factors in development of hyperplastic obesity. Am J Physiol 1998, 275: R1898-R1908.

19. Tang QQ, Otto TC, Lane MD: Commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci U S A 2004, 101: 9607-9611.

20. Sul HS, Smas C, Mei B, Zhou L: Function of pref-1 as an inhibitor of adipocyte differentiation. Int J Obes Relat Metab Disord 2000, 24 Suppl 4: S15-S19.

21. Umek RM, Friedman AD, McKnight SL: CCAAT-enhancer binding protein: a component of a differentiation switch. Science 1991, 251: 288-292.

22. Altiok S, Xu M, Spiegelman BM: PPARgamma induces cell cycle withdrawal: inhibition of E2F/DP DNA-binding activity via down-regulation of PP2A. Genes Dev 1997, 11: 1987-1998.

23. Pairault J, Green H: A study of the adipose conversion of suspended 3T3 cells by using glycerophosphate dehydrogenase as differentiation marker. Proc Natl Acad Sci U S A 1979, 76: 5138-5142.

24. Entenmann G, Hauner H: Relationship between replication and differentiation in cultured human adipocyte precursor cells. Am J Physiol 1996, 270: C1011-C1016.

25. MacDougald OA, Lane MD: Transcriptional regulation of gene expression during adipocyte differentiation. Annu Rev Biochem 1995, 64: 345-373.

26. Goldberg IJ: Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis. J Lipid Res 1996, 37: 693-707.

Page 146: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

132

27. Amri EZ, Dani C, Doglio A, Grimaldi P, Ailhaud G: Coupling of growth arrest and expression of early markers during adipose conversion of preadipocyte cell lines. Biochem Biophys Res Commun 1986, 137: 903-910.

28. Tontonoz P, Hu E, Spiegelman BM: Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell 1994, 79: 1147-1156.

29. Spiegelman BM, Ginty CA: Fibronectin modulation of cell shape and lipogenic gene expression in 3T3-adipocytes. Cell 1983, 35: 657-666.

30. Paulauskis JD, Sul HS: Cloning and expression of mouse fatty acid synthase and other specific mRNAs. Developmental and hormonal regulation in 3T3-L1 cells. J Biol Chem 1988, 263: 7049-7054.

31. Garcia dH, Birnbaum MJ: The acquisition of increased insulin-responsive hexose transport in 3T3-L1 adipocytes correlates with expression of a novel transporter gene. J Biol Chem 1989, 264: 19994-19999.

32. Bernlohr DA, Angus CW, Lane MD, Bolanowski MA, Kelly TJ, Jr.: Expression of specific mRNAs during adipose differentiation: identification of an mRNA encoding a homologue of myelin P2 protein. Proc Natl Acad Sci U S A 1984, 81: 5468-5472.

33. Jones BH, Standridge MK, Moustaid N: Angiotensin II increases lipogenesis in 3T3-L1 and human adipose cells. Endocrinology 1997, 138: 1512-1519.

34. Napolitano LM: Observations on the fine structure of adipose cells. Ann N Y Acad Sci 1965, 131: 34-42.

35. Weiner FR, Shah A, Smith PJ, Rubin CS, Zern MA: Regulation of collagen gene expression in 3T3-L1 cells. Effects of adipocyte differentiation and tumor necrosis factor alpha. Biochemistry 1989, 28: 4094-4099.

36. Kuri-Harcuch W, Arguello C, Marsch-Moreno M: Extracellular matrix production by mouse 3T3-F442A cells during adipose differentiation in culture. Differentiation 1984, 28: 173-178.

37. Aratani Y, Kitagawa Y: Enhanced synthesis and secretion of type IV collagen and entactin during adipose conversion of 3T3-L1 cells and production of unorthodox laminin complex. J Biol Chem 1988, 263: 16163-16169.

38. Ono M, Aratani Y, Kitagawa I, Kitagawa Y: Ascorbic acid phosphate stimulates type IV collagen synthesis and accelerates adipose conversion of 3T3-L1 cells. Exp Cell Res 1990, 187: 309-314.

39. Antras J, Hilliou F, Redziniak G, Pairault J: Decreased biosynthesis of actin and cellular fibronectin during adipose conversion of 3T3-F442A cells. Reorganization of the cytoarchitecture and extracellular matrix fibronectin. Biol Cell 1989, 66: 247-254.

Page 147: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

133

40. Ibrahimi A, Bonino F, Bardon S, Ailhaud G, Dani C: Essential role of collagens for terminal differentiation of preadipocytes. Biochem Biophys Res Commun 1992, 187: 1314-1322.

41. Fukai F, Iso T, Sekiguchi K, Miyatake N, Tsugita A, Katayama T: An amino-terminal fibronectin fragment stimulates the differentiation of ST-13 preadipocytes. Biochemistry 1993, 32: 5746-5751.

42. Ballas CB, Zielske SP, Gerson SL: Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem Suppl 2002, 38: 20-28.

43. Zandstra PW, Nagy A: Stem cell bioengineering. Annu Rev Biomed Eng 2001, 3: 275-305.

44. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR et al.: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002, 418: 41-49.

45. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ et al.: Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001, 7: 211-228.

46. Guilak F, Lott KE, Awad HA, Cao Q, Hicok KC, Fermor B et al.: Clonal analysis of the differentiation potential of human adipose-derived adult stem cells. J Cell Physiol 2006, 206: 229-237.

47. Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K et al.: Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem 2004, 14: 311-324.

48. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H et al.: Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002, 13: 4279-4295.

49. Yu G, Wu X, Dietrich MA, Polk P, Scott LK, Ptitsyn AA et al.: Yield and characterization of subcutaneous human adipose-derived stem cells by flow cytometric and adipogenic mRNA analyzes. Cytotherapy 2010, 12: 538-546.

50. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE et al.: Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 2004, 109: 1292-1298.

51. Planat-Benard V, Silvestre JS, Cousin B, Andre M, Nibbelink M, Tamarat R et al.: Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation 2004, 109: 656-663.

Page 148: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

134

52. Sengenes C, Lolmede K, Zakaroff-Girard A, Busse R, Bouloumie A: Preadipocytes in the human subcutaneous adipose tissue display distinct features from the adult mesenchymal and hematopoietic stem cells. J Cell Physiol 2005, 205: 114-122.

53. Cao Y, Sun Z, Liao L, Meng Y, Han Q, Zhao RC: Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochem Biophys Res Commun 2005, 332: 370-379.

54. Patrick CW, Jr., Chauvin PB, Hobley J, Reece GP: Preadipocyte seeded PLGA scaffolds for adipose tissue engineering. Tissue Eng 1999, 5: 139-151.

55. Kral JG, Crandall DL: Development of a human adipocyte synthetic polymer scaffold. Plast Reconstr Surg 1999, 104: 1732-1738.

56. Fischbach C, Spruss T, Weiser B, Neubauer M, Becker C, Hacker M et al.: Generation of mature fat pads in vitro and in vivo utilizing 3-D long-term culture of 3T3-L1 preadipocytes. Exp Cell Res 2004, 300: 54-64.

57. Halbleib M, Skurk T, de Luca C, von Heimburg D, Hauner H: Tissue engineering of white adipose tissue using hyaluronic acid-based scaffolds. I: in vitro differentiation of human adipocyte precursor cells on scaffolds. Biomaterials 2003, 24: 3125-3132.

58. von Heimburg D, Zachariah S, Heschel I, Kuhling H, Schoof H, Hafemann B et al.: Human preadipocytes seeded on freeze-dried collagen scaffolds investigated in vitro and in vivo. Biomaterials 2001, 22: 429-438.

59. Flynn L, Prestwich GD, Semple JL, Woodhouse KA: Adipose tissue engineering in vivo with adipose-derived stem cells on naturally derived scaffolds. J Biomed Mater Res A 2009, 89: 929-941.

60. Huss FR, Kratz G: Mammary epithelial cell and adipocyte co-culture in a 3-D matrix: the first step towards tissue-engineered human breast tissue. Cells Tissues Organs 2001, 169: 361-367.

61. Halberstadt C, Austin C, Rowley J, Culberson C, Loebsack A, Wyatt S et al.: A hydrogel material for plastic and reconstructive applications injected into the subcutaneous space of a sheep. Tissue Eng 2002, 8: 309-319.

62. Patrick CW, Jr., Wu X: Integrin-mediated preadipocyte adhesion and migration on laminin-1. Ann Biomed Eng 2003, 31: 505-514.

63. Patel PN, Gobin AS, West JL, Patrick CW, Jr.: Poly(ethylene glycol) hydrogel system supports preadipocyte viability, adhesion, and proliferation. Tissue Eng 2005, 11: 1498-1505.

64. Tabata Y, Miyao M, Inamoto T, Ishii T, Hirano Y, Yamaoki Y et al.: De novo formation of adipose tissue by controlled release of basic fibroblast growth factor. Tissue Eng 2000, 6: 279-289.

Page 149: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

135

65. Kimura Y, Ozeki M, Inamoto T, Tabata Y: Time course of de novo adipogenesis in matrigel by gelatin microspheres incorporating basic fibroblast growth factor. Tissue Eng 2002, 8: 603-613.

66. Masuda T, Furue M, Matsuda T: Photocured, styrenated gelatin-based microspheres for de novo adipogenesis through corelease of basic fibroblast growth factor, insulin, and insulin-like growth factor I. Tissue Eng 2004, 10: 523-535.

67. Patrick CW, Jr., Zheng B, Johnston C, Reece GP: Long-term implantation of preadipocyte-seeded PLGA scaffolds. Tissue Eng 2002, 8: 283-293.

68. Sosnik A, Sefton MV: Semi-synthetic collagen/poloxamine matrices for tissue engineering. Biomaterials 2005, 26: 7425-7435.

69. Gale NW, Yancopoulos GD: Growth factors acting via endothelial cell-specific receptor tyrosine kinases: VEGFs, angiopoietins, and ephrins in vascular development. Genes Dev 1999, 13: 1055-1066.

70. Patel ZS, Mikos AG: Angiogenesis with biomaterial-based drug- and cell-delivery systems. J Biomater Sci Polym Ed 2004, 15: 701-726.

71. Kimura H, Weisz A, Kurashima Y, Hashimoto K, Ogura T, D'Acquisto F et al.: Hypoxia response element of the human vascular endothelial growth factor gene mediates transcriptional regulation by nitric oxide: control of hypoxia-inducible factor-1 activity by nitric oxide. Blood 2000, 95: 189-197.

72. Eliceiri BP, Paul R, Schwartzberg PL, Hood JD, Leng J, Cheresh DA: Selective requirement for Src kinases during VEGF-induced angiogenesis and vascular permeability. Mol Cell 1999, 4: 915-924.

73. Conway EM, Collen D, Carmeliet P: Molecular mechanisms of blood vessel growth. Cardiovasc Res 2001, 49: 507-521.

74. Carmeliet P: Fibroblast growth factor-1 stimulates branching and survival of myocardial arteries: a goal for therapeutic angiogenesis? Circ Res 2000, 87: 176-178.

75. Lindahl P, Bostrom H, Karlsson L, Hellstrom M, Kalen M, Betsholtz C: Role of platelet-derived growth factors in angiogenesis and alveogenesis. Curr Top Pathol 1999, 93: 27-33.

76. Djonov V, Schmid M, Tschanz SA, Burri PH: Intussusceptive angiogenesis: its role in embryonic vascular network formation. Circ Res 2000, 86: 286-292.

77. Cao Y: Adipose tissue angiogenesis as a therapeutic target for obesity and metabolic diseases. Nat Rev Drug Discov 2010, 9: 107-115.

78. Friedman JM, Halaas JL: Leptin and the regulation of body weight in mammals. Nature 1998, 395: 763-770.

Page 150: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

136

79. Powell K: Obesity: the two faces of fat. Nature 2007, 447: 525-527.

80. Crossno JT, Jr., Majka SM, Grazia T, Gill RG, Klemm DJ: Rosiglitazone promotes development of a novel adipocyte population from bone marrow-derived circulating progenitor cells. J Clin Invest 2006, 116: 3220-3228.

81. Tang W, Zeve D, Suh JM, Bosnakovski D, Kyba M, Hammer RE et al.: White fat progenitor cells reside in the adipose vasculature. Science 2008, 322: 583-586.

82. Cao R, Brakenhielm E, Wahlestedt C, Thyberg J, Cao Y: Leptin induces vascular permeability and synergistically stimulates angiogenesis with FGF-2 and VEGF. Proc Natl Acad Sci U S A 2001, 98: 6390-6395.

83. Dobson DE, Kambe A, Block E, Dion T, Lu H, Castellot JJ, Jr. et al.: 1-Butyryl-glycerol: a novel angiogenesis factor secreted by differentiating adipocytes. Cell 1990, 61: 223-230.

84. Scroyen I, Jacobs F, Cosemans L, De Geest B, Lijnen HR: Effect of plasminogen activator inhibitor-1 on adipogenesis in vivo. Thromb Haemost 2009, 101: 388-393.

85. Hotamisligil GS: Inflammation and metabolic disorders. Nature 2006, 444: 860-867.

86. Nakagami H, Morishita R, Maeda K, Kikuchi Y, Ogihara T, Kaneda Y: Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy. J Atheroscler Thromb 2006, 13: 77-81.

87. Traktuev DO, Merfeld-Clauss S, Li J, Kolonin M, Arap W, Pasqualini R et al.: A population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circ Res 2008, 102: 77-85.

88. Suga H, Matsumoto D, Eto H, Inoue K, Aoi N, Kato H et al.: Functional implications of CD34 expression in human adipose-derived stem/progenitor cells. Stem Cells Dev 2009, 18: 1201-1210.

89. Fukumura D, Ushiyama A, Duda DG, Xu L, Tam J, Krishna V et al.: Paracrine regulation of angiogenesis and adipocyte differentiation during in vivo adipogenesis. Circ Res 2003, 93: e88-e97.

90. Crandall DL, Busler DE, McHendry-Rinde B, Groeling TM, Kral JG: Autocrine regulation of human preadipocyte migration by plasminogen activator inhibitor-1. J Clin Endocrinol Metab 2000, 85: 2609-2614.

91. Merfeld-Clauss S, Gollahalli N, March KL, Traktuev DO: Adipose tissue progenitor cells directly interact with endothelial cells to induce vascular network formation. Tissue Eng Part A 2010, 16: 2953-2966.

Page 151: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

137

92. Castellot JJ, Jr., Karnovsky MJ, Spiegelman BM: Differentiation-dependent stimulation of neovascularization and endothelial cell chemotaxis by 3T3 adipocytes. Proc Natl Acad Sci U S A 1982, 79: 5597-5601.

93. Hutley LJ, Herington AC, Shurety W, Cheung C, Vesey DA, Cameron DP et al.: Human adipose tissue endothelial cells promote preadipocyte proliferation. Am J Physiol Endocrinol Metab 2001, 281: E1037-E1044.

94. Varzaneh FE, Shillabeer G, Wong KL, Lau DC: Extracellular matrix components secreted by microvascular endothelial cells stimulate preadipocyte differentiation in vitro. Metabolism 1994, 43: 906-912.

95. Katz AJ, Llull R, Hedrick MH, Futrell JW: Emerging approaches to the tissue engineering of fat. Clin Plast Surg 1999, 26: 587-603, viii.

96. Carmeliet P, Jain RK: Angiogenesis in cancer and other diseases. Nature 2000, 407: 249-257.

97. Patrick CW, Jr.: Adipose tissue engineering: the future of breast and soft tissue reconstruction following tumor resection. Semin Surg Oncol 2000, 19: 302-311.

98. Frerich B, Lindemann N, Kurtz-Hoffmann J, Oertel K: In vitro model of a vascular stroma for the engineering of vascularized tissues. Int J Oral Maxillofac Surg 2001, 30: 414-420.

99. Lee KY, Peters MC, Anderson KW, Mooney DJ: Controlled growth factor release from synthetic extracellular matrices. Nature 2000, 408: 998-1000.

100. Yoon YS, Johnson IA, Park JS, Diaz L, Losordo DW: Therapeutic myocardial angiogenesis with vascular endothelial growth factors. Mol Cell Biochem 2004, 264: 63-74.

101. Neubauer M, Fischbach C, Bauer-Kreisel P, Lieb E, Hacker M, Tessmar J et al.: Basic fibroblast growth factor enhances PPARgamma ligand-induced adipogenesis of mesenchymal stem cells. FEBS Lett 2004, 577: 277-283.

102. Neubauer M, Hacker M, Bauer-Kreisel P, Weiser B, Fischbach C, Schulz MB et al.: Adipose tissue engineering based on mesenchymal stem cells and basic fibroblast growth factor in vitro. Tissue Eng 2005, 11: 1840-1851.

103. Grothe C, Nikkhah G: The role of basic fibroblast growth factor in peripheral nerve regeneration. Anat Embryol (Berl) 2001, 204: 171-177.

104. Tabata Y, Yamada K, Hong L, Miyamoto S, Hashimoto N, Ikada Y: Skull bone regeneration in primates in response to basic fibroblast growth factor. J Neurosurg 1999, 91: 851-856.

Page 152: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

138

105. Fujisato T, Sajiki T, Liu Q, Ikada Y: Effect of basic fibroblast growth factor on cartilage regeneration in chondrocyte-seeded collagen sponge scaffold. Biomaterials 1996, 17: 155-162.

106. Vashi AV, Abberton KM, Thomas GP, Morrison WA, O'Connor AJ, Cooper-White JJ et al.: Adipose tissue engineering based on the controlled release of fibroblast growth factor-2 in a collagen matrix. Tissue Eng 2006, 12: 3035-3043.

107. Butler MJ, Sefton MV: Poly(butyl methacrylate-co-methacrylic acid) tissue engineering scaffold with pro-angiogenic potential in vivo. J Biomed Mater Res A 2007, 82: 265-273.

108. Chen WY, Abatangelo G: Functions of hyaluronan in wound repair. Wound Repair Regen 1999, 7: 79-89.

109. Hung WS, Fang CL, Su CH, Lai WF, Chang YC, Tsai YH: Cytotoxicity and immunogenicity of SACCHACHITIN and its mechanism of action on skin wound healing. J Biomed Mater Res 2001, 56: 93-100.

110. Tanaka Y, Tsutsumi A, Crowe DM, Tajima S, Morrison WA: Generation of an autologous tissue (matrix) flap by combining an arteriovenous shunt loop with artificial skin in rats: preliminary report. Br J Plast Surg 2000, 53: 51-57.

111. Cronin KJ, Messina A, Knight KR, Cooper-White JJ, Stevens GW, Penington AJ et al.: New murine model of spontaneous autologous tissue engineering, combining an arteriovenous pedicle with matrix materials. Plast Reconstr Surg 2004, 113: 260-269.

112. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N et al.: Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Circulation 2002, 106: 3009-3017.

113. Menasche P, Hagege AA, Scorsin M, Pouzet B, Desnos M, Duboc D et al.: Myoblast transplantation for heart failure. Lancet 2001, 357: 279-280.

114. Phillips MI, Tang YL, Pinkernell K: Stem cell therapy for heart failure: the science and current progress. Future Cardiol 2008, 4: 285-298.

115. Madonna R, De Caterina R: Adipose tissue: a new source for cardiovascular repair. J Cardiovasc Med (Hagerstown ) 2010, 11: 71-80.

116. Chamberlain MD, Gupta R, Sefton MV: Chimeric vessel tissue engineering driven by endothelialized modules in immunosuppressed Sprague-Dawley rats. Tissue Eng Part A 2010.

117. Nor JE, Peters MC, Christensen JB, Sutorik MM, Linn S, Khan MK et al.: Engineering and characterization of functional human microvessels in immunodeficient mice. Lab Invest 2001, 81: 453-463.

Page 153: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

139

118. Skovseth DK, Yamanaka T, Brandtzaeg P, Butcher EC, Haraldsen G: Vascular morphogenesis and differentiation after adoptive transfer of human endothelial cells to immunodeficient mice. Am J Pathol 2002, 160: 1629-1637.

119. Nor JE, Christensen J, Mooney DJ, Polverini PJ: Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am J Pathol 1999, 154: 375-384.

120. Schechner JS, Nath AK, Zheng L, Kluger MS, Hughes CC, Sierra-Honigmann MR et al.: In vivo formation of complex microvessels lined by human endothelial cells in an immunodeficient mouse. Proc Natl Acad Sci U S A 2000, 97: 9191-9196.

121. Enis DR, Shepherd BR, Wang Y, Qasim A, Shanahan CM, Weissberg PL et al.: Induction, differentiation, and remodeling of blood vessels after transplantation of Bcl-2-transduced endothelial cells. Proc Natl Acad Sci U S A 2005, 102: 425-430.

122. Saunders WB, Bohnsack BL, Faske JB, Anthis NJ, Bayless KJ, Hirschi KK et al.: Coregulation of vascular tube stabilization by endothelial cell TIMP-2 and pericyte TIMP-3. J Cell Biol 2006, 175: 179-191.

123. Tremblay PL, Hudon V, Berthod F, Germain L, Auger FA: Inosculation of tissue-engineered capillaries with the host's vasculature in a reconstructed skin transplanted on mice. Am J Transplant 2005, 5: 1002-1010.

124. Koike N, Fukumura D, Gralla O, Au P, Schechner JS, Jain RK: Tissue engineering: creation of long-lasting blood vessels. Nature 2004, 428: 138-139.

125. Lin CS, Xin ZC, Deng CH, Ning H, Lin G, Lue TF: Defining adipose tissue-derived stem cells in tissue and in culture. Histol Histopathol 2010, 25: 807-815.

126. Miranville A, Heeschen C, Sengenes C, Curat CA, Busse R, Bouloumie A: Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 2004, 110: 349-355.

127. Planat-Benard V, Silvestre JS, Cousin B, Andre M, Nibbelink M, Tamarat R et al.: Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation 2004, 109: 656-663.

128. Borges J, Mueller MC, Padron NT, Tegtmeier F, Lang EM, Stark GB: Engineered adipose tissue supplied by functional microvessels. Tissue Eng 2003, 9: 1263-1270.

129. Traktuev DO, Prater DN, Merfeld-Clauss S, Sanjeevaiah AR, Saadatzadeh MR, Murphy M et al.: Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells. Circ Res 2009, 104: 1410-1420.

130. Niechajev I, Sevcuk O: Long-term results of fat transplantation: clinical and histologic studies. Plast Reconstr Surg 1994, 94: 496-506.

Page 154: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

140

131. Mauney JR, Nguyen T, Gillen K, Kirker-Head C, Gimble JM, Kaplan DL: Engineering adipose-like tissue in vitro and in vivo utilizing human bone marrow and adipose-derived mesenchymal stem cells with silk fibroin 3D scaffolds. Biomaterials 2007, 28: 5280-5290.

132. Feng Z, Yamato M, Akutsu T, Nakamura T, Okano T, Umezu M: Investigation on the mechanical properties of contracted collagen gels as a scaffold for tissue engineering. Artif Organs 2003, 27: 84-91.

133. Sosnik A, Cohn D, San Roman J, Abraham GA: Crosslinkable PEO-PPO-PEO-based reverse thermo-responsive gels as potentially injectable materials. J Biomater Sci Polym Ed 2003, 14: 227-239.

134. Sosnik A, Leung B, McGuigan AP, Sefton MV: Collagen/poloxamine hydrogels: cytocompatibility of embedded HepG2 cells and surface-attached endothelial cells. Tissue Eng 2005, 11: 1807-1816.

135. O'Connor KC, Song H, Rosenzweig N, Jansen DA: Extracellular matrix substrata alter adipocyte yield and lipogenesis in primary cultures of stromal-vascular cells from human adipose. Biotechnol Lett 2003, 25: 1967-1972.

136. Flynn L, Semple JL, Woodhouse KA: Decellularized placental matrices for adipose tissue engineering. J Biomed Mater Res A 2006, 79: 359-369.

137. Ades EW, Candal FJ, Swerlick RA, George VG, Summers S, Bosse DC et al.: HMEC-1: establishment of an immortalized human microvascular endothelial cell line. J Invest Dermatol 1992, 99: 683-690.

138. Santiago LY, Nowak RW, Peter RJ, Marra KG: Peptide-surface modification of poly(caprolactone) with laminin-derived sequences for adipose-derived stem cell applications. Biomaterials 2006, 27: 2962-2969.

139. Vashi AV, Keramidaris E, Abberton KM, Morrison WA, Wilson JL, O'Connor AJ et al.: Adipose differentiation of bone marrow-derived mesenchymal stem cells using Pluronic F-127 hydrogel in vitro. Biomaterials 2008, 29: 573-579.

140. McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS: Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev Cell 2004, 6: 483-495.

141. Cho SW, Kim SS, Rhie JW, Cho HM, Choi CY, Kim BS: Engineering of volume-stable adipose tissues. Biomaterials 2005, 26: 3577-3585.

142. McGuigan AP, Sefton MV: Design and fabrication of sub-mm-sized modules containing encapsulated cells for modular tissue engineering. Tissue Eng 2007, 13: 1069-1078.

Page 155: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

141

143. Weidner N, Semple JP, Welch WR, Folkman J: Tumor angiogenesis and metastasis--correlation in invasive breast carcinoma. N Engl J Med 1991, 324: 1-8.

144. Marxen M, Thornton MM, Chiarot CB, Klement G, Koprivnikar J, Sled JG et al.: MicroCT scanner performance and considerations for vascular specimen imaging. Med Phys 2004, 31: 305-313.

145. Akhtar N, Dickerson EB, Auerbach R: The sponge/Matrigel angiogenesis assay. Angiogenesis 2002, 5: 75-80.

146. Nehls V, Drenckhahn D: A novel, microcarrier-based in vitro assay for rapid and reliable quantification of three-dimensional cell migration and angiogenesis. Microvasc Res 1995, 50: 311-322.

147. Dietrich F, Lelkes PI: Fine-tuning of a three-dimensional microcarrier-based angiogenesis assay for the analysis of endothelial-mesenchymal cell co-cultures in fibrin and collagen gels. Angiogenesis 2006, 9: 111-125.

148. Pfaffl MW: A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001, 29: e45.

149. Nakatsu MN, Sainson RC, Aoto JN, Taylor KL, Aitkenhead M, Perez-del-Pulgar S et al.: Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and Angiopoietin-1. Microvasc Res 2003, 66: 102-112.

150. Neels JG, Thinnes T, Loskutoff DJ: Angiogenesis in an in vivo model of adipose tissue development. FASEB J 2004, 18: 983-985.

151. Straczkowski M, Dzienis-Straczkowska S, Stepien A, Kowalska I, Szelachowska M, Kinalska I: Plasma interleukin-8 concentrations are increased in obese subjects and related to fat mass and tumor necrosis factor-alpha system. J Clin Endocrinol Metab 2002, 87: 4602-4606.

152. Loskutoff DJ, Samad F: The adipocyte and hemostatic balance in obesity: studies of PAI-1. Arterioscler Thromb Vasc Biol 1998, 18: 1-6.

153. Orban JM, Wilson LB, Kofroth JA, El Kurdi MS, Maul TM, Vorp DA: Crosslinking of collagen gels by transglutaminase. J Biomed Mater Res A 2004, 68: 756-762.

154. Engler AJ, Sen S, Sweeney HL, Discher DE: Matrix elasticity directs stem cell lineage specification. Cell 2006, 126: 677-689.

155. Rowlands AS, George PA, Cooper-White JJ: Directing osteogenic and myogenic differentiation of MSCs: interplay of stiffness and adhesive ligand presentation. Am J Physiol Cell Physiol 2008, 295: C1037-C1044.

Page 156: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

142

156. Benoit DS, Schwartz MP, Durney AR, Anseth KS: Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nat Mater 2008, 7: 816-823.

157. Fumimoto Y, Matsuyama A, Komoda H, Okura H, Lee CM, Nagao A et al.: Creation of a rich subcutaneous vascular network with implanted adipose tissue-derived stromal cells and adipose tissue enhances subcutaneous grafting of islets in diabetic mice. Tissue Eng Part C Methods 2009, 15: 437-444.

158. Arts CH, Heijnen-Snyder GJ, Joosten PP, Verhagen HJ, Eikelboom BC, Sixma JJ et al.: A novel method for isolating pure microvascular endothelial cells from subcutaneous fat tissue ideal for direct cell seeding. Lab Invest 2001, 81: 1461-1465.

159. Hutley LJ, Herington AC, Shurety W, Cheung C, Vesey DA, Cameron DP et al.: Human adipose tissue endothelial cells promote preadipocyte proliferation. Am J Physiol Endocrinol Metab 2001, 281: E1037-E1044.

Page 157: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

143

9 Appendix 1:  HMEC Harvesting 

The human dermal microvascular endothelial cell line (HMEC-1) was used as the EC

source in Chapter 3 because they show the characteristic morphology and protein expression of

primary human HMEC while requiring less stringent growth medium and can be passaged

indefinitely. While cell lines may be useful for preliminary experiments, they have been altered

and as such are not phenotypically equivalent to primary cells. The use of primary cells was

necessary for the experiments in Chapter 4 and 5 of this thesis. HMEC dervived from the dermis

is available commercially (Cambrex, VEC) but we feel that EC derived from the same fat source

as the ASC may have an advantage. Harvesting both the HMEC and ASC from a single tissue

sample, we can envision an engineered fat pad created entirely from the patient’s own cells. This

would overcome many of the immune/rejection issues associated with cell transplantation.

Review of the literature revealed that magnetic separation of the cells using anti-body

binding to paramagnetic beads was required to obtain a pure population of endothelial

cells[158,159]. Even with magnetic separation, obtaining a pure population of endothelial cells

is a challenge because any contaminating adipose precursor cells will quickly overgrow any

existing endothelial cell population. We employed the MiniMACS™ cell separation columns

using degradable magnetic beads bound with platelet-endothelial cell adhesion molecule 1

(PECAM-1) (Miltenyi Biotech.)

Cells were harvested from freshly excised adipose tissue as per ASC harvest protocol in

section 3.3.1. which contains a mixture of both ASC and HMEC. Table 3 highlights the

experiments, conditions and results of the cell separation. Cells obtained from the harvest were

either pre-plated in T-150 flasks or directly separated at Passage 0 (P0) using either 1 column or

2 columns in series (to improve separation efficiency). Cells were then cultured in T-75 flasks

Page 158: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

144

and were separated again at Passage 1 (P1). All trials that cultured the cells in ‘coculture media’

were found to have complete overgrowth of ASC at Passage 2. The ‘coculture media’ was

developed by a previous student in the lab and contains the ASC base media (DMEM-F12

Ham’s, 10% FBS) with additional factors EGF, hydrocortisone and L-glutamine to support

HMEC-1 proliferation. This media may not support the attachment/proliferation of primary

HMEC or promote rapid proliferation of contaminating ASC, making separation difficult. For

trail 6 and 7, MCDB-131 (VEC Technologies) complete media was used which is recommended

for primary HMEC culture and gave positive results. It was decided to separate the cells directly

from harvest as pre-plating was found to only give the ASC cells time to overgrow the HMEC

population. Trial 7, using 2 columns in series during each separation, was successful and yielded

a nearly pure population of HMEC. Light microscopy images revealed cells with a typical

cobblestone morphology similar to the HMEC-1 and characteristic of endothelial cells.

(contaminating ASC in Trial 6 can be observed as long, spindle-shaped cells which were not

apparent in Trial 7) (Figure 9-1). Immunostaining for vonWillebrand’s factor (vWF) (green) on

cells prepared by cytospin from Trial 6 and 7 confirmed the presence of endothelial cells (Figure

9-2). Many non-stained cells are observed in Trial 6, while using 2 columns in series at each

separation was key to obtaining a nearly pure culture of HMEC in Trial 7. Several successful

separations using the conditions of Trial 7 have been conducted with reproducible results.

Page 159: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

145

Table 3: HMEC Separation Trials

Trial Method Media P0 (# columns) P1 (# columns)

P2 (result)

1 Harvest coculture 1 1 ASC overgrowth

2 Preplating (1.5h) coculture 1 1 ASC overgrowth

3 Preplating (72h) coculture 1 1 ASC overgrowth

4 Harvest coculture 2 2 ASC overgrowth

5 Preplating (72h) coculture 2 2 ASC overgrowth

6 Harvest MCDB 131 1 1 ASC contamination

7 Harvest MCDB 131 2 2 EC cobblestone morphology

Figure 9-1: Light microscopy images of cells obtained from HMEC separation trials.

Light microscopy images of cells obtained from Trial 6, and 7 and HMEC-1. Cobblestone morphology of cells in Trial 7, similar to HMEC-1, is indicative of endothelial cells. ASC contamination is observed in Trial 6 with long, spindle-like cells present. Scale bar = 100µm.

HMEC-1 Trial 7 – 2 Columns Trial 6 – 1 Column

Page 160: Modular Approach to Adipose Tissue Engineering...ii Modular Approach to Adipose Tissue Engineering Mark James Butler Doctor of Philosophy Department of Chemical Engineering and Applied

146

Figure 9-2: Cytospin slides of cells from Trial 6 and 7 immunostained with vWF

Most cells in Trial 7 stain positive for vWF (green) indicating a homgeneous population of HMEC. Trial 6 cells are mostly unstained contaminating ASC. Hoescht stains cell nuclei blue.

Trial 6 – 1 column

Trail

Trial 7 – 2 columns