pjm university de 10 sherbrooke developpement d'un … · pjm university de 10 sherbrooke...

134
PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT D'UN SYSTEME TRIDIMENSIONNEL DE CULTURE CELLULAIRE POUR ORIENTER LA FORMATION DE MICROVAISSEAUX DEVELOPMENT OF A TRIDIMENSIONAL CELL CULTURE SYSTEM TO ORIENT MICRO VESSEL FORMATION These de doctorat Speciality : genie chimique Irza SUKMANA Jury : Professeur Patrick VERMETTE (Directeur) Professeur Pierre PROULX (Rapporteur) Professeur Jean-Francois BEAULIEU Professeur Elizabeth JONES S herbrooke (Quebec) Canada Mai 2010 y^D^l

Upload: buihanh

Post on 19-Aug-2019

215 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

PJM UNIVERSITY DE

10 SHERBROOKE Faculte de genie

Departement de genie chimique et de genie biotechnologique

DEVELOPPEMENT D'UN SYSTEME TRIDIMENSIONNEL DE CULTURE CELLULAIRE

POUR ORIENTER LA FORMATION DE MICROVAISSEAUX

DEVELOPMENT OF A TRIDIMENSIONAL CELL CULTURE SYSTEM TO ORIENT

MICRO VESSEL FORMATION

These de doctorat Speciality : genie chimique

Irza SUKMANA

Jury : Professeur Patrick VERMETTE (Directeur) Professeur Pierre PROULX (Rapporteur) Professeur Jean-Francois BEAULIEU Professeur Elizabeth JONES

S herbrooke (Quebec) Canada Mai 2010

y^D^l

Page 2: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

1*1 Library and Archives Canada

Published Heritage Branch

395 Wellington Street OttawaONK1A0N4 Canada

Bibliotheque et Archives Canada

Direction du Patrimoine de I'edition

395, rue Wellington Ottawa ON K1A 0N4 Canada

Your file Votre reference ISBN: 978-0-494-70629-9 Our file Notre reference ISBN: 978-0-494-70629-9

NOTICE: AVIS:

The author has granted a non­exclusive license allowing Library and Archives Canada to reproduce, publish, archive, preserve, conserve, communicate to the public by telecommunication or on the Internet, loan, distribute and sell theses worldwide, for commercial or non­commercial purposes, in microform, paper, electronic and/or any other formats.

L'auteur a accorde une licence non exclusive permettant a la Bibliotheque et Archives Canada de reproduire, publier, archiver, sauvegarder, conserver, transmettre au public par telecommunication ou par I'lnternet, prefer, distribuer et vendre des theses partout dans le monde, a des fins commerciales ou autres, sur support microforme, papier, electronique et/ou autres formats.

The author retains copyright ownership and moral rights in this thesis. Neither the thesis nor substantial extracts from it may be printed or otherwise reproduced without the author's permission.

L'auteur conserve la propriete du droit d'auteur et des droits moraux qui protege cette these. Ni la these ni des extraits substantiels de celle-ci ne doivent etre imprimes ou autrement reproduits sans son autorisation.

In compliance with the Canadian Privacy Act some supporting forms may have been removed from this thesis.

Conformement a la loi canadienne sur la protection de la vie privee, quelques formulaires secondares ont ete enleves de cette these.

While these forms may be included in the document page count, their removal does not represent any loss of content from the thesis.

Bien que ces formulaires aient inclus dans la pagination, il n'y aura aucun contenu manquant.

1+1

Canada

Page 3: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

As a milestone of our 13 anniversary I dedicate this work to my wife,

the other half of my soul Ika Nury Wahidah.

To my mother and the memory of my late father (Almarhum) To my beloved son and daughters

To brothers and sisters.

Page 4: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

RESUME

Le developpement de micro-vaisseaux fonctionnels dans la culture de substituts tissulaires

constitue un enorme defi du genie tissulaire et de la medecine regeneratrice. Celui-ci est

justifie par la necessite de permettre une meilleure alimentation en nutriments et oxygene des

tissus cultives et une bonne elimination des dechets a l'interieur de ceux-ci. C'est la raison

pour laquelle seulement quelques substituts de tissus sont disponibles pour des applications

medicales. Par consequent, de nombreuses etudes de recherche dans le domaine de l'ingenierie

tissulaire ont mis l'accent sur la culture de micro-vaisseaux a l'interieur de matrices tissulaires

et ce avant leur implantation, tel que presente dans le Chapitre 1 de cette these. Le Chapitre 2

est consacre a l'etude bibliographique presentant revolution recente et les defis futurs lies au

processus de micro-vascularisation de substituts tissulaires. Ce processus est souvent appele

pre-vascularisation.

Le travail experimental presente dans cette these est base sur l'hypothese que des mono­

filaments polymeriques separes les uns des autres d'une facon precise peuvent etre utilises

pour guider des cellules endotheliales dispersees dans un gel de fibrine et ainsi induire des

structures tubulaires de facon directionnelle.

Le Chapitre 3 de cette these presente la validation de la formation de micro-vaisseaux dans un

environnement tridimensionnel. La methode de culture tissulaire presentee ici utilise des fibres

de polymere pour guider des cellules endotheliales et ce afin d'orienter la formation des

micro-vaisseaux. La methode d'attachement cellulaire decrite au Chapitre 3 a permis

l'adherence et la migration de cellules endotheliales humaines de la veine ombilicale

(HUVEC) sur des fibres de poly(ethylene terephtalate) (PET) non-modifiees. Les fibres

recouvertes de HUVEC ont ete « sandwichees » entre deux gels de fibrine pour permettre le

developpement des micro-vaisseaux. Le developpement de l'angiogenese a ensuite ete

caracterise a l'aide d'un certain nombre de techniques d'imagerie, y compris la microscopie par

fluorescence et la microscopie confocale. Apres 4 jours de culture, les micro-vaisseaux

formaient de grandes structures en forme de tube (diametre d'environ 100|im) entre les fibres

adjacentes distancees de 0.1mm.

ii

Page 5: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Le Chapitre 4 presente d'un cote l'etude de l'effet des fibroblastes humains et, d'un autre cote,

l'influence de deux facteurs de croissance angiogeniques (i.e., Vascular Endothelial Growth

Factor (VEGF) et le basic Fibroblast Growth Factor (bFGF)) sur les reponses des HUVEC

recouvrant les fibres de PET cultivees en sandwich dans des gels de fibrine. Le developpement

et la maturation des micro-vaisseaux ainsi que la formation des lumens ont ete etudies par

microscopie confocale en utilisant une matrice de fibrine fluorescente et par histologic

L'histologie et les experiences menees avec la fibrine fluorescente ont confirme que ces

pseudo-micro-vaisseaux formes entre les fibres de polymere incorporees dans la fibrine

avaient un lumen. Les effets du VEGF et du bFGF etaient fonction de la concentration de ces

molecules. L'utilisation de fibroblastes a considerablement ameliore la maturation des micro-

vaisseaux comparativement aux echantillons controles et ceux cultives avec le VEGF et le

bFGF.

La conclusion et les propositions pour des travaux fiiturs constituent le Chapitre 5.

Les Annexe A et B presentent, respectivement, les protocoles detailles utilises dans cette etude

pour marquer les cellules et des informations generates sur le cytosquelette.

Mots-cles: polyethylene terephtalate; orientation cellulaire; angiogenese directionnelle;

micro-vaisseaux; lumens multi-cellulaires; cultures en bioreacteur.

HI

Page 6: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

ABSTRACT The development of functional and oriented microvessels within tissue constructs is a

tremendous challenge in tissue engineering and regenerative medicine. It is justified by the

need to allow better nutrient and oxygen supply and waste removal within the core of tissue

constructs. It is one of the reasons why only few tissue substitutes are available to clinicians.

Therefore, the focus of many research studies in the field of tissue engineering has been placed

on promoting microvessel ingrowth inside tissue constructs prior to their implantation, as

presented in Chapter 1. This process is often referred to as pre-vascularization. As such,

Chapter 2 of this thesis is devoted to review the recent development and future challenges

related to the micro vascularization process of tissue constructs.

The experimental work in this thesis is based on the hypothesis that polymer monofilaments

precisely distanced from each others can be used to guide endothelial cells when dispersed in a

(fibrin) gel and to induce tube-like structures in a directional fashion.

In Chapter 3 of this thesis, the use of polymer fibres as contact guidance of endothelial cells to

orient microvessel formation and development in a three-dimensional environment was

validated. The novel cell attachment method described in Chapter 3 has resulted in an increase

in Human Umbilical Vein Endothelial Cell (HUVEC) adhesion and spreading on bare

poly(ethylene terephthalate) (PET) fibres. Furthermore, HUVEC-covered fibres were

sandwiched between fibrin gels to allow the development of microvessels. Angiogenesis

development was characterized and evaluated using a number of imaging techniques,

including fluorescence microscopy and confocal microscopy. After 4 days of culture,

microvessels formed large tube-like structures (diameter of approximately 100 um) between

adjacent fibres distanced by 0.1mm. This proof-of-concept opens the door to other

experiments and to possibility of scaling the system to bioreactor cultures.

In Chapter 4, the effect of human fibroblasts and in another set of experiments, the influence

of two angiogenic growth factors (i.e., Vascular Endothelial Growth Factor (VEGF) and basic

Fibroblast Growth Factor (bFGF)) over the responses of the HUVEC-covered fibres

IV

Page 7: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

sandwiched in fibrin were investigated. The development and maturation of microvessels as

well as the assessment of lumen formation were evaluated using a fluorescent fibrin matrix,

confocal microscopy and histology. Histology and fluorescent fibrin experiments confirmed

that these microvessel-like structures formed between polymer monofilaments embedded in

fibrin contained a lumen. The effects of VEGF and bFGF were dose dependent. Furthermore,

the use of fibroblasts significantly improved the maturation of the microvessels compared to

control and to samples cultured with VEGF and bFGF.

Conclusions and suggested future work are presented in Chapter 5.

Appendices A and B present the detailed protocols used to label cells in this study and general

information about cell cytoskeleton, respectively.

Keywords: Polyethylene terephthalate; Contact guidance; Cell patterning; Directional

angiogenesis; Microvessel formation; multi-cellular lumen; bioreactor cultures.

v

Page 8: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Acknowledgements

First of all, I would like to express my sincere gratitude to my supervisor Professor Patrick

Vermette, whose enthusiastic support and good advices helped me to go through my

challenging project.

I am very honoured to have Professor Pierre Proulx and Professor Jean-Francois Beaulieu for

their guidance and for being my pre-Doctoral examiners.

A very special thanks to Mr. Yannick Laplante and Dr. Aftab Ahamed, former members of the

Laboratoire de Bioingenierie et de Biophysique de l'Universite de Sherbrooke, for their

precious help when I started my Ph.D. I thank them also for their hints on general technical

and scientific aspects.

I also want to thank all my friends in the group for sharing their ideas and for scientific

discussions on daily basis during this course.

I should thank all the technicians in the Department of Chemical and Biotechnological

Engineering, especially Mr. Mark Couture for his help in designing and producing frames and

culture chambers for my research. Also, I want to thank Mr. Gilles Grondin and Dr. Leonid

Volkov for their contribution in confocal microscopy and Mrs. Johanne Dussault and Dr.

Sameh Geha who helped me with histology.

I am particularly thankful to my sponsor, the Islamic Development Bank (IDB), for all

financial support and living guidance for me and my family.

I am also very grateful to the Muslim community in Sherbrooke who has given me an

invaluable support and help in countless ways. I will never forget the support provided by dear

brothers: Hendra Hermawan, Abdelfettah and Rachid Bannari, Brahim Selma, Mbark El

vi

Page 9: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Morsli, Rachid Kadouche, Taha Abd-El-Rahman, Ahmad Kamal, Hamdy and Habibie, Deyab

El-Gamal, Ahmed Omran, Said and Hoccain and others, "Jazakumullahu khairan katsiran".

Finally, my most acknowledgment goes to my adorable wife, adik Ika Nury Wahidah, and our

lovely kids: Muhammad, Wafa and Sky. The word "Thanks" is never enough for their love,

patience and encouragement. May Allah SWT give you all the best rewards. Amen.

vn

Page 10: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

TABLE OF CONTENT

ABSTRACT ii

RESUME iv

Acknowledgements vi

Table of Content viii

List of Figures xi

List of Tables xii

List of Abbreviations xiii

CHAPTER 1 Introduction 1

1.1 Thesis Objectives 1

1.2 Thesis Structure 1

1.3 Contributions 3

CHAPTER 2 Vascularization strategies to engineer tissue substitutes: a challenge to support

blood microvessel development 5

2.1 Abstract 6

2.2 Introduction 6

2.3 Scaffolds in tissue engineering 8

2.3.1 Synthetic polymers 10

2.3.2 Natural polymers 11

2.4 Material properties 21

2.4.1 Scaffold porosity 21

2.4.2 Matrix stiffness 23

2.5 Scaffold vascularisation 24

2.5.1 Vasculogenesis and angiogenesis 28

2.5.1.1 Model 1: Cell death and phagocytosis 29

2.5.1.2 Model 2: Wrapping spaces around ECM 29

2.5.1.3 Model 3: Vacuole formation from the coalescence of intracellular vacuoles 31

2.5.2 Promoting angiogenesis in three-dimensional constructs 31

2.5.2.1 Micro-patterning 32

via

Page 11: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2.5.2.2 Surface modification 33

2.5.2.3 ECM modification 34

2.5.3 Co-culture systems and microvessel maturation 35

2.6 Bioreactor to support vascularization 37

2.7 Concluding remarks 39

CHAPTER 3 Polymer fibres as contact guidance to orient microvascularization in a 3D

Environment 57

3.1 Abstract 58

3.2 Introduction 58

3.3 Materials and methods 59

3.3.1 Materials 59

3.3.2 Cell culture : 60

3.3.3 PET fibres and fibrin gel preparation 60

3.3.4 Cell adhesion assay 61

3.3.5 Cell culture in 3D environment 61

3.3.6 Visualization of microvessels 62

3.3.7 Images and image processing 62

3.3.8 Statistical analysis 63

3.3.9 Confocal microscopy imaging of microvessels 63

3.4 Results 64

3.4.1 Cell adhesion on untreated PET fibres 64

3.4.2 Effect of fibre-to-fibre distance over cell connections and microvessel formation 65

3.4.3 Microvessel formation 66

3.5 Discussion 67

3.6 Conclusions 69

CHAPTER 4 The effects of co-culture with fibroblasts and angiogenic growth factors on

microvascular maturation and multi-cellular lumen formation in HUVEC-oriented

polymer fibre constructs 82

4.1 Abstract.... 83

4.2 Introduction 83

4.3 Materials and methods 85

ix

Page 12: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

4.3.1 Materials 85

4.3.2 Cell culture 85

4.3.3 Culture system and fibrin preparation 86

4.3.4 Cell adhesion and angiogenesis assay 86

4.3.5 Visualization of microvessel and lumen formation 87

4.3.6 Histological section 88

4.3.7 Imaging microvessels by confocal microscopy 88

4.3.8 Statistical analysis 89

4.4 Results and discussion 89

4.5 Conclusions 93

CHAPTER 5 Conclusions and suggestions 107

5.1 Conclusions 107

5.2 Conclusion (en francais) 108

5.3 Suggestions 109

5.3.1 Further studies of the angiogenesis assessment 109

5.3.2 The effect of dynamic culture system over microvessel development and proof of

microvessel functionality 111

APPENDIX A Protocol for cell staining 116

APPENDIX B Cell cytoskeleton 119

x

Page 13: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

LIST OF FIGURES

Figure 2.1: Fibrinogen structure and fibrin clot process 16

Figure 2.2: Models of the development of tube-like structures during vasculogenesis 30

Figure 2.3: Blood vessel structure and development 36

Figure 3.1: Schematic illustration of the in vitro method designed to guide and orient

microvessel formation 75

Figure 3.2: HUVEC attachment on PET fibres 76

Figure 3.3: Effect of fibre-to-fibre distance over cell connections and microvessel

development 77

Figure 3.4: Confocal and inverted microscopy image 78

Figure 3.5: Schematic of the microvascularization process 79

Supplementary Figure 3.i: Scanning electron microscopy (SEM) image of a PET fibre 80

Supplementary Figure 3.ii: Progression of cell connections and microvessel reorganization

along and in between adjacent fibres sandwiched between two fibrin gels 81

Figure 4.1: Effect of fibroblasts over microvessel development 100

Figure 4.2: Immunofluorescence picture illustrating the effect of fibroblasts on microvessel

development 101

Figure 4.3: Immunofluorescence picture (F-actin and nuclei staining) illustrating the effect of

VEGF and bFGF on microvessel development 102

Figure 4.4: Illustration of multi-cellular lumen formation for cultures carried out for a period

of 4 days with fibroblasts 103

Supplementary Figure 4.i: Comparison between systems using cell-free fibres (left) and

HUVEC-covered fibres (right) over cell-to-cell connections & microvessel

development 104

Supplementary Figure 4.ii: Immunofluorescence confocal pictures of a sample cultured with

no fibroblasts 105

Supplementary Figure 4.iii: Fibroblasts stained with the CFSE CellTracer 106

Figure 5.1: Simple set-up for dynamic cell cultures 113

XI

Page 14: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

LIST OF TABLES

Table 2.1: Integrins and non-integrin receptors which can bind to fibrin(ogen) 18

Table 2.2: The use of synthetic and natural polymers to support tissue vascularisation 19

Table 3.1: Effects of fibre-to-fibre distance on cells 74

Table 4.1: Effects of VEGF and bFGF concentrations over the number of cells and cell-cell

connections following 2 days of culture 98

Table 4.2: Comparison of the effects of fibroblasts, VEGF, and bFGF over the number of

cells and cell-cell connections 98

Table 4.3: Review of some studies on the effects of VEGF and bFGF on endothelial cells and angiogenesis 99

xn

Page 15: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

LIST OF ABBREVIATIONS

3D

Agp-1 ANOVA

BAEC bFGF

BSA CAM CLS CSTR

DLLA EC ECGS

ECM ePTFE

ERG

ESAM

FBS FDA FGF

GAG HA HBSS HDMEC

HUVEC

HVEC

ICAM-1

MC

three-dimensional or tridimensional angiopoietin-1 analysis of variance

bovine aortic endothelial cell basic fibroblast growth factor

bovine serum albumin chorioallontoic membrane capillary-like structure continuous stirred-tank reactor (D,L-lactic acid) endothelial cell endothelial cell growth supplement extracellular matrix expanded poly(tetrafluoroethylene) early response genes

endothelial cell selective adhesion molecule foetal bovine serum Food and drug administration fibroblast growth factor

glycosaminoglycan hyaluronic acid hanks balances salt saline human dermal endothelial cell human umbilical vein endothelial cell human vascular endothelial cell inter-cellular adhesion molecule-1 messenchymal cell

o-HA

PBS PDGF-BB PDLA PECAM-1 PEG PET PGA PLA

PLG PLGA PLLA

RAOEC RGD

RHAMM

SEM

SF SMC SM-MHC SMocA STLV TMC VE-cadherin VEGF

VSMC

vWF

oligosaccharides of HA

phosphate buffer saline platelet-derived growth factor-BB poly(D,L-lactic acid) platelet endothelial cell adhesion molecule-1 poly(ethylene glycol) poly(ethylene terephthalate) poly(glycolic acid) poly(lactic acid)

poly(lactic glycolic) poly(lactic-co-glycolic acid) poly(L-lactic acid)

rat aortic endothelial cell arginine-glycine-aspartic acid peptide receptor for HA mediated motility scanning electron microscopy skin fibroblast smooth muscle cell smooth muscle myosin heavy chain smooth mucle a-actin slow turning lateral vessel trimethylene carbonate vascular endothelial cadherin

vascular endothelial growth factor vascular smooth muscle cell

von Willebrand factor

X1I1

Page 16: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

CHAPTER 1

Introduction The development of functional and oriented microvessels within engineered human tissue

substitutes constitutes a promising hope in tissue engineering and regenerative medicine

[Stock and Vacanti 2001; Eichmann et al. 2005]. To date, the development of thick tissues,

such as pancreas, liver, heart, and kidneys is problematic due to the lack of construct

vascularisation, resulting in cell and tissue death [Atala 2004; Jain et al. 2005; Kannan et al.

2005]. The focus of current research efforts in tissue engineering has been mainly on

developing strategies to study and promote microvascularization within tissue constructs

[Kannan et al. 2005; Neuman et al. 2003].

1.1 Thesis objectives

This project was designed to constitute the initial part of a larger research program aiming to

create an oriented functional blood microvessel network within three-dimensional tissue

scaffolds. One of the aims of this research program is to scale-up tissue engineering methods

by the use of dynamic culture conditions using an automated bioreactor system. The principal

objective of this thesis was to design and validate static cell culture processes to guide

endothelial cell responses and the subsequent microvessel development and orientation.

1.2 Thesis Structure

This thesis has been written in a manner that chapters correspond to scientific articles resulting

from this work. At the time of submission, two articles were published and one under

preparation for submission. Below is a brief description of their content.

Chapter 1: Introduction. This section contains thesis objectives and structure, and

contributions.

1

Page 17: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Chapter 2: Vascularization strategies to engineer tissue substitutes: a challenge to

support blood microvessel development. This chapter is a literature review on advancements

in the field of three-dimensional tissue engineering with an emphasis on cellular and

angiogenesis guidance as well as on blood microvessel development. Moreover, the effects of

cell types, co-cultures, and growth factors on microvessel development are discussed. The use

of bioreactors to culture tissue substitutes is also briefly addressed.

Chapter 3: Polymer fibers as contact guidance to orient microvascularization in a 3D

environment. This part of the thesis presents a novel culture system for angiogenesis

guidance using poly(ethylene terephthalate) (PET) fibres. Briefly, in an effort to increase the

bioactivity of PET fibres, cells are first allowed to attach and cover the fibres using a novel

culture method. Then, HUVEC-covered fibres are sandwiched in fibrin and microvessel

structures are examined by confocal microscopy. In addition, the effect of fibre-to-fibre

distance over angiogenesis development is compared. Results from this chapter reveal that this

culture system can be used to orient microvessel structures. The optimum fibre-to-fibre

distance found here is further used in the subsequent part of this thesis.

Chapter 4: The effect of co-culture with fibroblasts and angiogenic growth factors on

microvascular maturation and multi-cellular lumen formation in HUVEC-oriented

polymer fibre constructs. This chapter further validates the use of the culture system

developed in Chapter 3 using a co-culture method with human skin fibroblasts or with

angiogenic growth factors (i.e., Vascular Endothelial Growth Factor (VEGF) and Basic

Fibroblast Growth Factor (bFGF)). The effects of these two parameters over microvessel

development and maturation are investigated. This study is also aiming to determine whether

or not these microvessels contained multi-cellular lumen. VEGF and bFGF are shown to

enhance HUVEC proliferation and migration, thus improving the development of cell-cell

connections and subsequent microvessel development. The use of fibroblasts results in more

mature microvessels containing more cell-cell connections, which also contain lumens.

Chapter 5: Conclusions and Suggestions. This part presents a general conclusion and

suggests future direction.

2

Page 18: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

1.3 Contributions

During the course of this thesis, I have had important help from several people, to whom I am

very grateful. Firstly, former members of the Laboratoire de Bioingenierie et de Biophysique

de l'Universite de Sherbrooke, Mr. Yannick Laplante, Dr. Aftab Ahamed, and Dr. Mbark El

Morsli, have helped me to gain technical know-how and scientific knowledge.

Mr. Yannick Laplante has taught me how to prepare protocols and techniques to carry out cell

culture. Furthermore, Mr. Laplante has assisted me in the preparation and evaluation of three-

dimensional culture systems. Dr. Aftab Ahamed has tough me statistics and how to prepare a

scientific article. Dr. Mbark El Morsli has shared his knowledge in advance mathematics and

chemical engineering.

The confocal microscopy was carried out at the Department of Biology (Universite de

Sherbrooke) and at the Centre de recherche clinique Etienne-Le Bel (Centre hospitalier

universitaire de Sherbrooke (CHUS)), respectively, where Mr. Gilles Grondin and Dr. Leonid

Volkov have shared knowledge and helped me to obtain good quality pictures for my

publications. Mrs Johanne Dussault and Dr. Sameh Geha have assisted me with histology.

After fixation, all my histology samples were sent to the Pathology Department (CHUS). The

sectioning and staining have been done by Mrs Johanne Dussault, while further expert

interpretation have been done by Dr. Sameh Geha.

All other laboratory and analytical works were carried out by me.

3

Page 19: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

References

Atala, A. (2004) Tissue engineering and regenerative medicine: concepts for clinical

application. Rejuvenation Res. 7, p. 15-31.

Eichmann, A., Noble, F. L., Autiero, M. and Carmeliet, P. (2005) Guidance of vascular and

neural network formation. Current Opinion in Neurobiology 15, p. 108-15.

Jain, R. K., Au, P., Tarn, J., Duda, D. G. and Fukumura, D. (2005) Engineering vascularized

tissue. Nat. Biotechnol. 23, p. 821-23.

Kannan, R. Y., Salacinski, H. J., Sales, K., Butler, P. and Seifalian, A. M. (2005) The roles of

tissue engineering and vascularisation in the development of micro-vascular networks: a

review. Biomaterials 26, p. 1857-75.

Neuman, T., Nicholson, B. S. and Sanders, J. E. (2003) Tissue engineering of perfused

microvessels. Microvasc. Res. 66, p. 59-67.

Stock, U. and Vacanti, J. P. (2001) Tissue engineering: current state and prospects. Annu. Rev.

Med. 52, p. 443-51.

4

Page 20: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

CHAPTER 2

Vascularization strategies to engineer tissue substitutes:

a challenge to support blood microvessel development

Irza Sukmana1'2

1. Laboratoire de Bioingenierie et de Biophysique de l'Universite de Sherbrooke, Department

of Chemical and Biotechnological Engineering, Universite de Sherbrooke, 2500, blvd de

l'Universite, Sherbrooke, QC, Canada, J1K 2R1.

2. Research Centre on Aging, Institut universitaire de geriatrie de Sherbrooke, 1036, rue

Belvedere Sud, Sherbrooke, QC, Canada, J1H 4C4.

5

Page 21: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2.1 Abstract

The guidance of endothelial cell organization into a capillary network has been a long standing

challenge in tissue engineering. Some research efforts have been made to develop methods to

promote blood capillary networks inside engineered tissue constructs. Mature microvessels

that would mimic blood microvessel function can be used to subsequently facilitate oxygen

and nutrient transfer as well as waste removal. Microvascularization of engineered tissue-

scaffold appears to be one of the most favorable approaches to overpass nutrient and oxygen

supply limitation, which is often the major hurdle in developing thick and complex tissue or

organ substitutes. This review chapter addresses recent advances and future challenges in

developing and using bioactive scaffolds and 3D culture systems to promote tissue construct

vascularization allowing mimicking blood microvessel development and function encountered

in vivo. Vascularization can be achieved through strategies in which endothelial cells are

cultured in bioactive scaffolds and/or extracellular matrix (ECM)-like environments, with or

without angiogenic growth factors as well as using cell co-cultures and combinations of those.

In the near future, bioreactors will be used to create fully vascularized functional tissue and

organ substitutes.

2.2 Introduction

Tissue engineering is an emerging field. In 1987, the National Science Foundation defined

tissue engineering as "an interdisciplinary field that applies the principles of engineering and

the life sciences towards the development of biological substitutes that restore, maintain or

improve tissue function" [Langer and Vacanti 2003]. Tissue engineering involve the repair

and restoration of various tissue and organ functions, while limiting host rejection and side

effects for the patient by delivering cells and/or biomolecules through the use of 3D scaffolds

[Fuchs et al. 2001; Mooney et al. 1997].

Although there have been some successes to replace and restore some tissues using tissue

engineering approaches, these have been mostly limited to thin or avascular tissues, such as

cartilage, skin or bladder [Mooney and Mikos 1999]. However, for thick and vascular tissues

and for most organs, the lack of a sufficient supply of nutrients and oxygen to growing tissues,

6

Page 22: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

as well as the waste removal represent two important factors that limit the successful

development and implantation of engineered tissue constructs [Arthur et al. 1998; Neumann et

al. 2003]. These issues of poor mass transport and mass transfer have often led to the failure of

the culture process or even to that of implants [Arthur et al. 1998; Sipe 2002].

One possible strategy for creating thick engineered tissue substitutes in vitro is to use a

bioactive scaffold that allows the guidance of endothelial cells promoting microvessel

development in a directional fashion in order to vascularized the tissue construct [Godbey and

Atala 2002]. It is quite well accepted among the scientific community that pre-vascularization

of tissue construct appears to be one of the most favorable and efficient approaches to address

the problem of tissue survival due to a lack of oxygen and nutrient supply [Atala 2004; Mikos

et al. 1993; Montano et al. 2009; Nomi et al. 2002]. The concept of pre-vascularization mainly

involves the incorporation of endothelial cells into a bioactive scaffold to form a capillary

network inside the structure prior to its implantation [Atala 2004; Langer et al. 1995]. This

could accelerate the formation of functional microvessels within the core of an implant

[Godbey and Atala 2002; Neumann et al. 2003].

The idea of using pre-vascularized engineered tissue substitutes was first suggested by Mikos

et al. (1993) when comparing the performance of pre-vascularized tissues to non-vascularized

ones. Later, Sakakibara et al. (2002) concluded that pre-vascularization enhanced the benefits

of cardiomyocyte transplantation. More recently, Levenberg et al. (2005) demonstrated that

pre-vascularization improved the performance of skeletal muscle tissue constructs when

implanted in mice. Furthermore, studies aiming to induce and control vascularization may also

advance general knowledge on the development of. therapeutics targeting angiogenesis.

Numerous pathological conditions are associated with insufficient oxygen and blood supply

[Atala 2004; Carmeliet 2005; Neumann et al. 2003]. Also, uncontrolled angiogenesis is

associated with many diseases, including rheumatoid arthritis, macular degeneration, and

tumour growth [Carmeliet and Conway 2001].

Advances in tissue engineering have brought significant knowledge on the mechanisms and

parameters related to vascularization and angiogenesis development and blood microvessel

7

Page 23: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

network formation [Levenberg and Langer 2004; Nerem 2006]. This review will report status

of the current research and development related to bioactive scaffolds and culture systems

designed to promote angiogenesis and vascularization. It is necessary to improve our

understanding of the mechanisms behind angiogenesis and to apply that knowledge to guide

microvessel growth in a regulated manner. Other aspects including the challenges in

angiogenesis guidance, assessment of angiogenesis and lumen formation and the use of

bioreactor system to culture vascularized tissue constructs will also be outlined.

2.3 Scaffolds in tissue engineering

The needs for engineered tissue substitutes are important. Currently, the demand for organ

transplants is higher than the supply. In the United States alone, there were 79,512 patients on

the transplantation waiting list in 2002, and only 24,422 received organs while 6,297 died

while waiting [Nerem 2006]. In addition, although organ transplantation is one of the less

expensive therapies in regenerative medicine, tissue engineering offers hope for more

consistent and rapid treatment of those in need [Lanza and Chick 1997].

As an interdisciplinary approach between engineering and life science, tissue engineering

seeks an opportunity to develop suitable biomaterial-cell hybrid constructs to support the

regeneration and restoration of tissue structure and function. The critical challenge facing

tissue engineering today relies on our knowledge and ability to fabricate tissue and organ

replacements that would fulfill physiological function [Koh et al. 2009; Nerem 2006]. Also,

the success of tissue engineering methods relies on the ability of the construct to integrate at

the implantation site with the native tissue. Tissue engineering is facing important clinical and

practical problems, such as cell sourcing, rejection, healing, and cell/tissue death [Conway and

Carmeliet 2004; Gimble and Guilak 2003].

Various key concepts in tissue engineering and regenerative medicine have been pursued to

overcome those problems and these concepts include injection of tissue-specific viable cells

directly into a damaged tissue (for example brain cells in the case of Parkinson or Alzheimer),

encapsulation of specific cell types within synthetic permeable matrices that allow release of

therapeutics (e.g., the release of insulin or dopamine from pancreatic islets in the treatment of

8

Page 24: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

diabetes), and to seed scaffolds with living cells in vitro, allowing their maturation before

being implanted [Lanza et al. 2000; Yang et al. 2001a]. In the present thesis, we are mainly

interested with the last concept.

If an isolated cell population can be expanded in vitro using cell culture and bioreactor

techniques, in theory, only a very small number of cells from donors would be necessary to

prepare such biological implants. Since the isolated cells cannot form new tissue by

themselves, a (temporary) template is needed, which we refer here to as scaffold. Scaffolds are

expected to provide a control over tissue architecture and mechanical properties. They can

allow cells to adhere, proliferate, and migrate [Howe et al. 1998; Huttenlocher et al. 1995] in

order to form a required structure and to synthesize their own extracellular matrix (ECM)

molecules [Chang and Werb 2001], thus hopefully allowing tissue regeneration or repair

[Nerem 2007; Yang et al. 2001b].

It is believed that the success to develop tissue constructs depends on many aspects, such as

cell sourcing, type of biomaterials used to make scaffolds, tissue culture methods, only to

name a few. For example, using cells from other species, such as pig, remains a debate since

there is a risk of transferring diseases from animals to humans [Bach 1998]. Using cells from

the same genotype or close relatives of a patient could avoid problems associated with

immune rejection, which can result in tissue death [Bach 1998; Lee et al. 2005; Ye et al.

2000b].

The behaviour of individual cells and the dynamic state of multi-cellular tissues are regulated

by the interaction between cells and their surrounding matrix. Therefore, the design of

scaffolds from the macroscopic scale (e.g., pore structure) to the nanoscopic level (e.g.,

surface properties) is very important. Firstly, decision of using either natural or synthetic

scaffolds should be based on their capability to provide a specific microenvironment that

mimics the natural environment of the targeted anatomical site [Garcia et al. 2005]. Secondly,

the three-dimensional scaffold should fulfill some requirements with respect to:

biocompatibility, degradation rate, porosity, mechanical properties (e.g., stiffness), and

chemistry (e.g., surface chemical/protein composition) [Chaikof et al. 2002].

9

Page 25: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In the in vivo environment, cells interact with their ECM in a dynamic manner. The concept of

dynamic "communication" between cells and their matrix has opened a wide exploration of

the ECM molecules and scaffold materials that can be used in tissue engineering. Scaffolds

can be made from synthetic polymers, naturally occurring materials, or a combination of both.

2.3.1 Synthetic polymers

Synthetic polymers have been investigated and used to make scaffolds in tissue engineering

for a variety of possible applications. The principal advantage of using synthetic polymers is

that their properties (e.g., biodegradation, physicochemistry and mechanical stiffness) can be

controlled by manipulating their molecular weight and compositions, for example [Chaikof et

al. 2002]. Among them, synthetic degradable polymers from the poly(a-esters) family, such as

poly(lactic acid) (PLA), poly(glycolic acid) (PGA) and their co-polymers, have been

extensively investigated in biomaterials and tissue engineering.

Synthetic polymers from the poly(a-esters) group are degraded mainly through chemical

hydrolysis and are mostly insensitive to enzymatic attack [Larson and Chu 2006], and often,

the degradation profile does not vary between patients [Mooney et al. 1997]. For example, it

was recognized that the constituting monomers of PLA and PGA are nontoxic and are

metabolized in the body [Tabata 2006; Tabata 2009]. Therefore, PLA, PGA and their

copolymers (e.g., poly(lactic-co-glycolic acid) (PLGA)) are FDA-approved and they can be

produced to form a variety of implants ranging from screws, meshes, and sutures to porous

scaffolds [Larson and Chu 2006; Lee et al, 2008; Tabata 2006].

PGA is an inelastic polyester with a high crystallinity (46-50%) and is degraded by water

(through hydrolysis) to form glycolic acid [Kikuchi et al. 1997; Vert et al. 1994]. PLA is less

crystalline, more hydrophobic, and less susceptible to hydrolysis than PGA [Larson III and

Chu 2006; Vert et al. 1994]. For tissue engineering applications, their copolymers, such as

PLLA (poly(L-lactic acid)), PLGA (poly(lactic-co-glycolic acid)) and PDLLA (poly(D,L-lactic

acid) are more popular, since their properties can be tailored [Vert et al. 1994]. For example,

the degradation time of PLLA has been reported to be very slow, while PDLLA hydrolysed in

a matter of weeks [Yang et al. 2001a]. Even a small amount of D,L-LA in the polymer chain of

10

Page 26: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

PLA can accelerate the degradation time dramatically [Bramfeld et al. 2007; Grizzi et al.

1995; Tabata 2006].

Several investigators have explored the potential use of polymers from the PLA and PGA

family to fabricate scaffolds for the promotion of microvascularization and angiogenesis. For

example, Grizzi et al. (1995) have produced poly(D,L-lactic-co-glicolic acid) scaffolds to

engineer tubular tissues. Furthermore, when endothelial cells were seeded in these constructs,

they were able to generate a capillary network inside the scaffold [Mooney et al. 1994;

Mooney et al. 1996]. Also, in a more recent study, Levenberg et al. (2005) have successfully

pre-vascularized PLLA/PLGA sponges with pore size ranging from 225 to 500um. Then, they

implanted the pre-vascularized scaffold in skeletal muscles of mice and found that the method

was able to promote angiogenesis in the implant.

In another application, D,L-lactic acid (DLLA) was combined with 1,3-trimethylene carbonate

(TMC) at a specific molecular weight ratio of 81:19 (DLLA:TMC) in order to produce a

TMC-DLLA copolymer. This copolymer was processed to make a scaffold with lOOum

average pore size having a high interconnectivity. This scaffold was tested in vitro and shown

to support cardiomyocyte survival [Pego et al. 2003b]. When this scaffold seeded with cells

was subcutaneously implanted in rats, it elicited an acute inflammatory reaction [Pego et al.

2003a].

2.3.2 Natural polymers

Comparing to synthetic polymers, natural polymers have longer history and have been broadly

used in many applications in the biomedical, pharmaceutical, and tissue engineering fields.

While synthetic scaffolds offer good mechanical properties and less product variability with a

high level of control, natural scaffolds provide a better environment for cell attachment and

signalling, resulting in more efficient regulation of cell structures and functions [Chiu et al.

2009; Grizzi et al. 1995]. Natural polymers can be made from proteins (e.g., collagens, gelatin,

albumin, and fibrinogen), polysaccharides (e.g., chitosan, hyaluronic acid, alginate, cellulose

and dextran), and their chemical derivatives.

11

Page 27: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

ECM-derived polymers are attractive materials to make bioactive scaffolds, since they can

provide cells with an environment more similar to the cell native ECM [Howe et al. 1998;

Larson III and Chu 2006]. The ECM can be defined as a complex protein structure outside the

cells, which mainly consists of collagens and proteoglycans. The primary function of the ECM

is to support the cellular structure. Some ECM components regulate cellular processes, such as

cell proliferation, motility, differentiation, migration and adhesion [Chiu et al. 2009].

Each tissue has a unique ECM composition and environment. Therefore, the design of ECM-

derived scaffold should mimic certain features and functions of the ECM for the targeted end

use. For example, in the case of scaffold vascularization, the matrix should provide an

environment such as in the connective tissue for the endothelial cells to adhere and proliferate

as well as to form and remodel vascular structures [Becker et al. 2009]. Furthermore, as

endothelial cells are lining the innermost layer of blood vessels and capillary microvessels,

their interaction with the underlying ECM is essential to maintain cellular integrity and

functional activity for the development of functional and mature blood vessels [Hutchings

2003; Jain 2003]. To date, natural polymers such as hyaluronic acid, chitosan, alginate,

collagens, and fibrin are the most important biodegradable materials to fabricate scaffolds.

Hyaluronic acid (HA), also known as hyaluronan, is a glycosaminoglycan (GAG) that has a

linear polysaccharide branch (glucuronic acid N-acetyl D-glucosamine). Hyaluronic acid is the

embryo's first ECM material and is present in nearly all adult mammalian tissues [Brekke et

al. 2006; Toole 2004]. Hyaluronic acid, with high molecular mass (ranging between 10 to

1,000 kDa), has a unique characteristic [Brekke et al. 2006; Slevin et al. 2002]. Indeed, HA

shows poor cell adherence and inhibits endothelial cell proliferation, while its degradation

products (e.g., oligosaccharides of HA, o-HA) are pro-angiogenic and, through chemotaxis,

can stimulate cell migration, differentiation and the overall angiogenesis process [Rao et al.

1997; Slevin et al. 2002]. For example, West et al. (1985) have demonstrated that o-HA

(molecular mass < 10 kDa) induced angiogenesis with human umbilical endothelial cells

(HUVEC) in the chorioallontoic membrane (CAM) assay. Furthermore, the CD44 receptors in

endothelial cells was found to bind o-HA and to initiate the expression of early response genes

(ERG), resulting in cell proliferation and migration [Lesley et al. 2000; Miletti-Gonzalez et al.

12

Page 28: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2005]. More recently, o-HA was reported to stimulate angiogenesis, either in vitro or in vivo,

with vascular endothelial cell through both CD44 and RHAMM (receptor for HA mediated

motility) [Lokeshwar and Selzer 2000; Miletti-Gonzalez et al. 2005; Slevin et al. 2002]. In

addition, added Fibroblast Growth Factor (FGF) in the HA matrix improved the

neovascularization of the construct [Rauh et al. 2006].

Other polysaccharides, such as chitosan and alginate, have also been investigated for various

tissue engineering and biomedical applications. Chitosan is a linear polysaccharide, composed

of iV-acetyl and D-glucosamine, and has been investigated for some applications, such as to

make contact lens [Park et al. 2003], matrix to encapsulate cells [Zielinski and Aebischer

1994], drug release device, and to engineer cartilage and bone substitutes [Rauh et al. 2006].

Chitosan is an attractive biomaterial to fabricate scaffolds [Park et al. 2003]. For example,

Black et al. (1999) and Slevin and Kumar (2002) have produced a scaffold made of chitosan

cross-linked with HA. This scaffold improved endothelial cell proliferation and induced

capillary network and angiogenesis development inside the construct [Black et al. 1999;

Slevin et al. 2002]. Alginate gel is a hydrogel that is not affected by temperature changes

[Draget et al. 1997]. It has been tested for drug delivery and cell transplantation [Draget et al.

1997; Drury and Mooney 2003; Smidsrod and Skjak-Braek 1990]. Furthermore, incorporating

vascular endothelial growth factor (VEGF) in alginate gels promoted neovascularization in the

matrix [Chang et al. 2010; Drury and Mooney 2003]. This system has been suggested as a

promising approach for clinical applications [Chang et al. 2010].

Collagens are the most abundant proteins, being found in nearly all tissues in mammalians

[Becker et al. 2009]. Type I, II, III, and IV are the most abundant forms and make up

approximately 90% of the collagens in the human body [Becker et al. 2009; Haarer and Dee

2006]. To date, over 25 types of collagens have been identified and have been processed into

various forms, including films, sponges, fibres, and gels [Haarer and Dee 2006]. Collagen type

I, II, III, V and XI can self-assemble into fibrils. Other collagens (e.g., type IV, VIII, and X)

form network and are found in the basement membrane [Becker et al. 2009; Chen et al. 1995].

13

Page 29: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In tissue engineering, physical and chemical cross-linking is often used to increase the

mechanical strength and to avoid rapid degradation of collagens [Chen et al. 1995]. Photo-

oxidation, de-hydrothermal treatment, and ultraviolet irradiation are examples of physical

cross-linking methods, while chemical methods include treatment with carbodiimides,

glutaraldehydes, and poly(glycidyl ether) [Ma et al. 2004]. Often, chemical methods result in

higher cross-linking degree; they are therefore more common than physical methods. Also,

chemical treatment parameters (e.g., time and temperature) as well as catalyst concentration

can be adapted to vary mechanical and degradation properties of collagen scaffolds [Angele et

al. 2004; Ma et al. 2004]. On the other hand, chemical methods could leave some potentially

toxic chemical residues [Ma et al. 2004].

Collagens can be purified from animal and human sources, but the concern of the

immunologic and disease transmissions, especially for animal collagens, still remains. To

avoid those risks, Toman et al. (1999) have suggested a method to produce recombinant

collagens. Recombinant human collagen types I and II are commercially available (e.g.,

FibroGen Inc, San Francisco, CA) [Becker et al. 2009; Toman et al. 1999]. Engineered tissue

substitutes for skin replacement called Apligraft™ (Organogenesis Inc, Canton,

Massachusetts, USA) is made from collagen type I and was the first commercialized man-

made tissue substitute [Zisch et al. 2003]. Collagen can also be extracted from tilapia

(Oreochromis niloticas). Indeed, Sugiura et al. (2009) have produced collagen sponge from

tilapia. In vivo implantation of the scaffold into rabbit muscle revealed that tilapia collagen

caused less inflammatory responses, when compared to porcine collagen [Sugiura et al. 2009].

Other studies have reported the use of type I and IV collagens to carry out angiogenesis and

vasculogenesis assays [Soker et al. 2000]. In vitro culture of endothelial cells in 3D matrix

made of type I collagen resulted in an increased number of tube-like structures and supported

angiogenesis development [Francis et al. 2008]. Also, with FGF, collagen type IV scaffold

supported endothelial cell growth and differentiation, thus regulating capillary development

[Ingber and Folkman 1989]. Xu et al. (2001) concluded that denaturation of collagen type IV

can promote a specific angiogenic cryptic epitope (i.e., HUIV26), which can bind to the

cellular integrin ctvP3. To date, at least four different collagen binding integrins on endothelial

14

Page 30: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

cells are known and these include ocipi, oc2pl, al0(3l, and a l i p i [Davis and Senger 2005;

Gulberg and Lundgren-Akerlund 2002; Senger et al. 1997; Xu et al. 2001].

Human fibrinogen is a large, complex and fibrous glycoprotein with a molecular weight of

340 kDa. It is 45nm in length and composed of two symmetric "D" domain molecules and a

central "E" domain. Each domain consists of one set of three different poly-peptide chains

termed Aa, Bp\ and y chain (see Figure 2.1.a) [Herrick et al. 1999; Tawil 2006]. In the body,

fibrinogen is present in human blood plasma at a concentration of approximately 2.5 g/L. The

protein is essential for haemostasis, wound healing, inflammation, angiogenesis, and other

biological events. Fibrinogen is a soluble macromolecule, which can be converted to an

insoluble gel (i.e., fibrin) to stabilize the haemostatic plug and to provide a temporary matrix

for subsequent cellular responses involved in wound healing [Janmey et al. 2009; Tawil 2006].

The role of fibrin in this process is not passive, but the protein rather actively directs cellular

responses through specific receptor-mediated interactions with blood cells (e.g., leucocytes) as

well as endothelial cells of the vessel wall [Herrick et al. 1999]. Therefore, the use of fibrin as

a bioactive scaffold to support tissue vascularization is of interest.

The formation of fibrin clot during wound healing is initiated by the release of thrombin, a

serine protease enzyme, which subsequently activates the coagulation cascade [Herrick et al.

1999; Rowe et al. 2007; Tawil 2006]. After the release, thrombin cleaves peptide fragments

from fibrinogen to generate the fibrin monomer (Fig. 2.1.b) by the clotting cascade into

protofibrils (Fig. 2.1.c). Afterward, in the presence of chloride ion and transglutaminase factor

XIII or factor XHIa, protofibrils undergo intermolecular cross-linking to form a stable fibrin

gel [Janmey et al. 2009; Litvinov et al, 2005; Rowe et al. 2007]. See Figure 2.1.d for details on

the fibrin formation as well as the degradation processes.

Changing fibrinogen or thrombin concentration can change the resulting fibrin material,

affecting both biochemical and mechanical properties [Ferrenq et al. 1997]. For example,

Vailhe et al. (1998) have shown that capillary-like structures made from HUVEC seeded on

fibrin depended on the mechanical factor of the gel. Harder gel, made using higher

concentration of fibrinogen, led to a decreased number of capillary-like structures. No

15

Page 31: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

capillary-like structures was found in softer matrix (< 0.5mg/mL of fibrinogen) as well as in

too rigid one (> 4mg/mL of fibrinogen) [Ferrenq et al. 1997; Vailhe et al. 1998].

a

/».*feY"^-'."% v chain

D domain E domain O domain « * * ** *

two A audi two & "kiwlis* IK ., }B T-nunluk • msH|Mk. ^ \ Ii ^ 0-m,vlnk jr-«i"lufc

"ibrm unworncr

|— [)-ii£i>iii—~-| —~— J -ivgj n —— j—~l)-rfjjttin

i>'«f»« D*ejih>fl protofibril

i"**,\. Am<mi €»**

c»-'

»»&*

P»'i*?IS»ril»

r-*Jt.» fasm *rt

Uteri*

Figure 2.1: Fibrinogen structure and fibrin clot process. Adapted from Herrick et al. (1999)

and Litvinov et al. (2005).

16

Page 32: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Increasing fibrinogen concentration can reduce the matrix pore size, thus hindering endothelial

cell migration and capillary formation [Nehls and Herrmann 1996]. In addition, Rowe et al.

(2007) found that decreasing thrombin concentration resulted in both an increased gel

compaction and micro-fibres size, thus causing different cellular morphology and alignment of

vascular smooth muscle cells. These examples illustrate that fibrin gel properties and

subsequent cell responses can be modulated, to some extent, opening the door to more

applications [Cox et al. 2004; Rowe et al. 2007].

To date, fibrin is commercially available as fibrin sealant or fibrin glue (e.g., Tisseel™, Baxter

AG, Vienna, Austria) for surgical applications [Tawil 2006]. Also, fibrin scaffolds have been

used in many tissue engineering applications, including as matrix to treat bone and skin

defects [Catelas et al. 2006], for drug delivery in neurological and cardiovascular disorders

[Ferguson et al. 2003; Tassipoulos et al. 2000], and for three-dimensional angiogenesis assays

[Vailhe et al. 1998; Nakatsu and Hughes 2008]. For example, Montesano et al. (1986) and

Dejana et al. (1987) found that fibrinogen induced adhesion, spreading, and microfilament

organization of human endothelial cells either in 2D or 3D in vitro culture system.

Also, culturing endothelial cells on microcarrier beads and then embedding these beads in

fibrin have been proposed by Nehls and Drenckhahn (1995). This system resulted in formation

of capillary structures and sprouting [Nehls and Drenckhahn 1995]. However, the system

failed to model sprouting angiogenesis containing multi-cellular lumen surrounded by

polarized endothelial cells, which is of importance during blood microvessel development

[Montano et al 2009; Carmeliet and Conway 2001; Yancopoulos et al. 2000]. More recent

vascularization studies using fibrin gels have been presented by Chen et al. (2009), Nakatsu et

al. (2003a), and Sukmana and Vermette (2010a). In our study (see Chapter 3 of this thesis),

endothelial cells proliferate and migrate along patterned polymer fibres and then fibrin is

degraded along with the formation of cell-cell interactions, leading to the formation of tube­

like structures, and eventually to sprouting and lumen formation with adjacent vessels

[Sukmana and Vermette 2010b].

17

Page 33: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Unlike synthetic hydrogels, fibrin is an active matrix for cells. It can bind many growth factors

and bioactive cloth components including fibronectin, hyaluronic acid, and von Willebrand

factor [Herrick et al. 1999; Weisel 2005]. Human fibrinogen can bind to endothelial cells

through either integrins or non-integrin binding sites (Table 2.1). For example, fibrin has two

pairs of RGD binding sites and a non-RGD site at the y chain, which can interact with

endothelial cell integrins (i.e., cc5pi, av(33, and anb(33) as well as with leucocyte integrins

(i.e., aMp3 and ccxp2) [Cheresh et al. 1989; Lishko et al. 2002; Tawil, 200; Weisel 2005]. The

location of integrins binding sites in fibrinogen is presented in Figure 2. La. Other non-integrin

receptors that can bind to endothelial cells include ICAM-1, CD-44 surface receptor, and

platelet endothelial cell adhesion molecule-1 (PECAM-1, also known as CD-31) [Cheresh et

al. 1989; Herrick et al. 1999; Monchaux and Vermette 2010].

Table 2.1: Integrins and non-integrin receptors which can bind to fibrin(ogen).

Integrins

ccvp3

oc5pl

ociibp3

ocMp3

«xP2

Non-integrin receptor

PECAM-1 (CD31)

ICAM-1

CD-44

Cell types

endothelial cells and fibroblasts

endothelial cells

platelets and endothelial cells

leucocytes and monocytes

lymphocytes and leucocytes

Cell types

endothelial cells

fibroblasts, endothelial cells

endothelial cells, fibroblasts and tumorous cells

Cellular function

adhesion and spreading

adhesion

adhesion and cloth retraction

adhesion and phagocytosis

adhesion

Cellular function

adhesion and von Willebrand factor release proliferation and adhesion

adhesion, migration, and proliferation

References

Cheresh et al. 1989; Cox et al. 2004; Monchaux and Vermette 2010. Cheresh et al. 1989; Herrick et al. 1999; Weisel 2005. Janmey et al. 2009; Monchaux and Vermette 2010. Herrick et al. 1999; Lishko et al 2002; Tawil 2006. Herrick et al. 1999; Janmey et al. 2009.

References

Monchaux and Vermette 2010; Nakatsu et al. 2003a; Cheresh et al. 1989; Herrick et al. 1999; Rowe et al. 2007. Herrick et al. 1999; Weisel 2005.

18

Page 34: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In other tissue engineering applications, fibrin was combined with collagen for the

development of blood vessel substitutes [Cummings et al. 2004; Stegemann et al. 2006].

Collagen type I is the predominant structural component of the media as well as the adventia

layers of blood vessels, while the inner layer of natural blood vessels, called the intima layer,

is lined by endothelial cells [Monchaux and Vermette 2010]. Therefore, the combination of

collagen-fibrin can be used to make scaffolds with good mechanical and biochemical

properties [Yao et al. 2005; Cummings et al. 2004]. For example, Isenberg et al. (2006) have

investigated a tubular scaffold made of type I collagen and fibrinogen to engineer small-

diameter artificial arteries.

A summary of the use of synthetic and natural polymers that have been applied to support

tissue vascularization is presented in Table 2.2.

Table 2.2: The use of synthetic and natural polymers to support tissue vascularization.

Scaffold Materials

Poly(D,L-lactic-co-glicolic acid)

PLLA/PLGA sponges

DLLA/TMC porous scaffold

Hyaluronic acid (HA)

HA cross-linked with chitosan

Purpose and Methods

• Scaffold designed for tubular tissues.

• EC were seeded in the construct.

• Scaffolds designed with pores from 225 to 500um for skeletal muscle tissue.

• Pre-seeded with MC, then scaffolds were implanted in skeletal muscles of mice.

• For heart tissue engineering with pore size oflOOum.

• Cardiomyocytes were seeded in the scaffold. Then scaffolds were subcutaneously implanted in rats.

Fibroblast growth factor (FGF) was added in the HA matrix.

Scaffold designed to produce a human skin

Results

EC were able to generate capillary network inside the scaffolds.

Scaffold pre-vascularization promoted angiogenesis in the implant.

This method elicited an acute inflammatory reaction.

FGF improved neovascularization.

The hybrid scaffold improved EC

References

Mooney et al. 1994; Mooney et al. 1996.

Levenberg et al. 2005.

Pego et al. 2003a; Pego et al. 2003b.

Rauh et al. 2006.

Black et al. 1999; Slevin et

19

Page 35: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Alginate gel

Marine collagen from tilapia (Oreochromis niloticas)

Collagen type I

Collagen type IV

Fibrin gel

equivalent. • Endothelial cells were

seeded in the construct.

• The construct was made for several applications including cell encapsulation.

• Vascular endothelial growth factor (VEGF) was incorporated in the scaffold.

Sponges were tested in vivo.

• In vitro angiogenesis assay.

• EC were seeded in the matrix.

Fibroblast growth factor (FGF) was incorporated in the scaffold for in vitro study of angiogenesis.

Fibrinogen and thrombin concentrations were changed to study angiogenesis, in vitro, using HUVEC and VSMC.

proliferation and induced al. 2002. capillary network formation inside the scaffold. VEGF promoted neovascularization.

Tilapia collagen caused less inflammatory responses when compared to porcine collagen. Type I of collagen increased the number of tube-like structures and supported angiogenesis development. • Addition of FGF in

type IV collagen scaffold supported EC growth and capillary formation.

• Degradation product of collagen type IV promoted a specific angiogenic epitope.

• Capillary-like structures (CLS) made of HUVEC depended on the matrix rigidity.

• Higher rigidity decreased CLS number.

• No CLS were found in very soft or very rigid matrices.

• Decreasing thrombin concentration caused different morphology and alignment of VSMC.

Chang et al. 2010; Drury and Mooney 2003.

Sugiura et al. 2009.

Francis et al. 2008; Soker et al. 2000.

Ingber and Folkman 1989; Xuetal. 2001.

Ferrenq et al. 1997; Nehls and Herrmann 1996; Rowe et al. 2007; Vailhe et al. 1998.

20

Page 36: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Fibrin gel in which microcarrier beads were embedded

Polymer monofilaments (i.e., PET) embedded in fibrin.

Fibrin gel combined with collagen type I

Aim: to generate angiogenesis development. Microcarrier beads were pre-coated with HUVEC and subsequently embedded in fibrin. In vitro study of angiogenesis guidance. PET fibres were pre-coated with HUVEC, then sandwiched in fibrin.

SMC were grown on a sheet-like scaffold, and then wrapped around tubular vessel. The vessel allowed to transfer nutrient and oxygen.

This in vitro culture system provided a step-by-step process of the capillary development containing multi-cellular lumen.

• Pre-coating PET with cells enabled increasing the fibres bioactivity.

• PET fibres were able to guide EC to orient microvessels.

This in vitro culture system enabled producing a better environment for the vascularization of small-caliber arterial substitutes.

Chen et al. 2009; Nakatsu et al. 2003a; Nehls and Drenckhahn 1995.

Sukmana and Vermette 2010a; Sukmana and Vermette 2010b.

Cummings et al. 2004; Isenberg et al. 2006; Yao et al. 2005.

EC: endothelial cell smooth muscle cell;

MC: mesenchymal cell; VSMC: vascular smooth muscle cell; SMC: PET: poly(ethylene terephthalate).

2.4 Material properties

Scaffold and ECM materials are selected based on bulk and surface properties, which can be

tuned with the aim to modulate cell adhesion and proliferation as well as phenotypic cell

expression [Francis et al. 2008]. Among the properties of importance, scaffold porosity and

matrix stiffness play significant roles in cell and tissue responses and these will be briefly

discussed below.

2.4.1 Scaffold porosity

Porosity is defined as the fraction of the void space over the total volume of a scaffold. Pore

size corresponds to the distance between two solid sections of the porous matrix [Lawrence et

al. 2008]. In tissue engineering, a highly porous scaffold (about 90% porosity) is more

desirable, since it should increase mass and nutrient transport [Kannan et al. 2005]. Higher

porosity can increase cell adhesion and provide a sufficient area for cell-matrix interactions

21

Page 37: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

and new ECM production by cells [Agrawal and Ray 2001; Kannan et al. 2005]. However,

bulk and mechanical properties should also be considered. At higher porosity, the total solid

volume of the solid part of the scaffold is lower, when compared to scaffolds with lower

porosity, thus resulting in weaker mechanical support [Kannan et al. 2005].

The effect of pore size over cell behaviour has been investigated in culturing bone tissue

substitutes. Pores in the range of 300-400um have been found to be optimal for osteoblast

attachment, growth and proliferation [Linnes et al. 2007]. The important role of such porous

structures in endothelial cell organization and angiogenesis development was pioneered by

Clowes et al. (1986) more than 20 years ago. In a more recent study, pore size was found to

have a significant effect over cell binding, morphology, and phenotype, thus inducing

endothelial cell migration and capillary formation inside the scaffold [O'Brien et al. 2005].

In the scientific literature, there are some suggestions concerning the optimum pore size to

support vascularization. For example, many mature cell types, including fibroblasts and

endothelial cells, have been found to be unable to spread and completely colonize the bulk of

scaffolds with pore size higher than 300um, due to the difficulty on bridging the distance

[Yanmas et al. 1989; Zeltinger et al. 2001]. In an in vitro angiogenesis study, it was shown

that a cell's ability to bridge the distance in 3D scaffolds is important to support the

vascularization process [Sukmana and Vermette 2010a]. Using HUVEC-covered

poly(ethylene terephthalate) (PET) monofilaments, as contact guidance in HUVEC-seeded

fibrin, it was suggested that the optimum fibre-fibre distance to support microvessel

development was lOOum [Sukmana and Vermette 2010a].

Furthermore, hepatocytes were also reported to spread well on gelatin-chitosan scaffold (3:1)

with pore size of 20um, while fibroblasts and endothelial cells spread better on the same

matrix but with pores ranging between 100-150um, compared to pores from 20-80um [Huang

et al. 2005]. The role of pore size to promote endothelial cell lining has been investigated in

vascular grafts. For example, Zhang et al. (2004) found that an external pore size of 30um was

preferable over 20um or smaller pores in terms of promoting rapid tissue ingrowth and

endothelial cell growth in the expanded poly(tetrafluoroethylene) (ePTFE) graft.

22

Page 38: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In addition, Marshall et al. (2004) found that fibrin with 35um pores significantly supported

angiogenesis development, when compared to fibrin with either 20um or 70um pores. Fibrin

with pore sizes of approximately 30um promotes the ingrowth of vascularized fibrous tissue in

engineered blood vessels [Harding et al. 2002]. However, the optimum porosity and pore size

of the scaffold is still open to question and is based on the application as well as the cell type

[Kannan et al. 2005; Zeltinger et al. 2001; Zhang et al. 2004].

2.4.2 Matrix stiffness

Substrate mechanical properties, such as stiffness, are known to be important parameters

affecting cell responses. In cell biology, matrix stiffness is sensed by cell receptors and

integrins transmit mechanical stress across the cell surface to the cell cytoskeleton (see

•Appendix B for a brief description), converting mechanical signals into (bio)chemical ones

[Becker et al. 2009; Geiger et al. 2001; Ingber et al. 1995].

Therefore, the ECM stiffness will influence cellular functions, including cell adhesion,

proliferation, migration as well as phenotype differentiation [Geiger et al. 2001; Ingber et al.

1995]. For example, Pelham and Wang (1997) have examined the effect of collagen-coated

poly(acrylamide) scaffold on the behaviour of rat epithelial and fibroblast cells. They found

that on more rigid (higher stiffness) surfaces, cells were more spread and showed increased

motility and focal adhesion contacts [Pelham and Wang 1997]. Furthermore, increasing

surface stiffness was found to result in increased cell contractility [Li et al. 2005], more

organized cells cytoskeleton and actin stress fibres, and higher adhesion strength [Discher et

al. 2005]. The phenomenon related to the effect of environment stiffness over cell behaviour is

known to as durotaxis [Lo et al. 2000].

Substrate stiffness and cell contractility also have a significant role over microvascular

development. For example, while endothelial cells proliferate more on rigid surfaces, they

form tube-like structures on softer substrates [Deroanne et al. 2001; Discher et al. 2005].

Vasculogenesis decreased with an increase in matrix stiffness, which was a result of an

increase in collagen [Angele et al. 2004; Haarer and Dee 2006; Yeung et al. 2005] or

fibrinogen [Duong et al. 2009; Tawil 2006; Vailhe et al. 1998] concentration.

23

Page 39: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2.5 Scaffold vascularization

Cells can be isolated from a patient or donor and seeded into a scaffold to allow cell

proliferation and support pre-vascularization process [Lanza et al. 2000; Mikos et al. 1993].

Subsequently, these constructs can be implanted in the same patient. After implantation,

hopefully endothelial cells would develop connections with the existing blood microvessels of

the surrounding tissue, forming a microvascular network and allowing adequate tissue

perfusion [Lanza et al. 2000; Ye et al. 2000a].

When tissues become thicker, cells located more than a few hundred microns (about 200 to

300um) from capillaries suffer from a lack of oxygen and nutrients, resulting into necrotic

conditions [Hutmatcher 2001; Ko et al. 2007]. Currently, the development of thick and

complex tissues and organs such as the heart, muscles, kidneys, liver and lung relies, in part,

on the knowledge and ability to stimulate microvascular network formation within tissue

constructs [Ko et al. 2007; Mooney and Mikos 1999]. Therefore, knowing how to carry out

cell seeding within a scaffold and to perform successful in vitro cultures to prevascularize

tissue constructs prior to their implantation is highly important. As such, researchers rely on

the increasing knowledge related to neovascularization (i.e., vasculogenesis and angiogenesis)

process to stimulate vascular network formation within three-dimensional tissue constructs

[Bramfeld et al. submitted-2010]. Before discussing vasculogenesis and angiogenesis

processes, I will highlight the cell types available and/or used to carry out these experiments.

The existence of capillaries in blood vessels and lung of frogs was first reported by Marcello

Malpighi in 1661 [Hillen et al. 2006]. After the invention of biological microscopes by

Antonie van Leeuwenhoek (1632-1727) and his colleagues and further development of

fluorescence techniques, the anatomy of blood vessels was revealed [Hillen et al. 2006]. Blood

vessels are composed of several cell types and the main ones are smooth muscle cells (SMC),

pericytes, fibroblasts and endothelial cells [Jain 2003; Ko et al. 2007].

Smooth muscle cells are found in blood vessels, such as in middle layer (i.e., tunica media) of

large and small blood vessels, in lymphatic vessels, uterus, and in the gastrointestinal and

respiratory systems [Hillen et al. 2006; Jain 2003]. Behind the basic function of vascular SMC

24

Page 40: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

in blood vessels i.e., to maintain vessels integrity and support the endothelium [Auerbach et al.

2003; Hillen et al. 2006], they are highly specialized cells for the regulation of blood vessel

diameter, vessel contraction, blood pressure and flow distribution [Auerbach et al. 2003;

Owens et al. 2004]. Furthermore, SMC synthesize the connective tissue matrix of the vessel

wall, which is composed of elastin, collagen, and proteoglycans [Auerbach et al. 2003; Hillen

et al. 2006]. SMC show a very low level of proliferation during normal development but

become more proliferative in response to vessel injury [Jain 2003].

There are some markers to identify smooth muscle cells, including smooth muscle cc-actin

(SMaA) [Owens et al. 2004], smooth muscle myosin heavy chain (SM-MHC) [Bramfeld et al.

submitted-2010; Hungerford and Little 1999], SM22cc, and calponin [Duband et al. 1993]. To

date, SMaA is the most commonly used marker of SMC, which represents up to 70% of the

actin population in vascular SMC [Bramfeld et al. submitted-2010; Hungerford and Little

1999; Jung etal. 2002].

Pericytes are perivascular-specific cells that are associated with capillaries and blood

microvessel development [Hillen 2006; Jain 2003]. Pericytes have the capacity to differentiate

into other cell types, including SMC, fibroblasts, and osteoblasts [Jung et al. 2002; Owens et

al. 2004]. During blood microvessel development, the coverage of capillary by pericytes is

important for the maturation, remodeling and maintenance of the vascular system via the

secretion of growth factors and/or modulation of the ECM [Allt and Lawrenson 2001; Jung et

al. 2002]. The important role of pericytes in capillary development in vitro has been studied by

some researchers. For example, endothelial cells co-cultured with pericytes, separated by a

cellulose membrane, resulted in inhibition of capillary growth, while when pericytes were in

contact or close to endothelial cells; endothelial cell growth and capillary development were

observed [Orlidge et al. 1987].

Fibroblasts are a cell type mostly found in connective tissues, including blood vessels,

cartilage, soft tissues (e.g., dermis) and bone. A connective tissue can be defined as a tissue

that wraps, connects, nourishes and supports all other tissues and organs [Becker et al. 2009].

Fibroblasts originate from mesenchymal cells - mesenchymal cells are progenitor cells

25

Page 41: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

capable of forming connective tissues and the lymphatic system [Auerbach et al. 2003; Jain

2003]. During normal development and wound healing process of connective tissues,

fibroblasts produce fibres and secrete factors to maintain the ECM and provide structural

support for the tissues [Auerbach et al. 2003]. To date, fibroblasts from human skin or skin

fibroblasts have been co-cultured with endothelial cells in many angiogenesis studies [Allt and

Lawrenson 2001; Chen et al. 2009; Nakatsu et al. 2003a; Sukmana and Vermette 2010b].

Also, 3T3 cells that were originally obtained from Swiss mouse embryo tissues have become a

standard fibroblast cell line [Allt and Lawrenson 2001]. In an in vitro study, skin fibroblasts

have been reported to have a significant effect over microvascular formation and angiogenesis

development from HUVEC [Nakatsu et al. 2003a; Sukmana and Vermette 2010b].

In the entire circulatory system, endothelial cells line the inner layer of the vessels (i.e., tunica

intima) and consist of more than 1013 cells for approximately 1kg of vessel [Jain 2003;

Sumpio et al. 2002]. The first culture of endothelial cells was reported by Shibuya around

1930, and then various techniques and sources of endothelial cells were investigated [Hillen et

al. 2006]. For in vitro studies, endothelial cells from animal or human sources can be used.

Endothelial cells from animals include canine jugular endothelial cells and aortic endothelial

cells either from bovine (BAEG), porcine (PAOEC) or rat (RAOEC) [Ko et al. 2007] sources.

Human endothelial cells include human umbilical vein (HUVEC), human dermal

microvascular (HDMEC) and human vascular (HVEC) [Tonello et al. 2003]. Since endothelial

cells from animals can result in immune responses if used in humans, and due to the available

sources of endothelial cells, HUVEC have been utilized by many research groups [Ko et al.

2007].

Heterogeneity between endothelial cells has been reported not only between large vessel-

derived cells and those of microvascular origin, but also between organs [Hillen et al. 2006;

Sumpio et al. 2002]. For example, aortic endothelial cells are thicker and cover a small area

compared to those from human pulmonary artery [Fuchs et al. 2001]. Also, endothelial cells

from microvascular (e.g., human dermal microvascular endothelial cells (HDMEC)) are

elongated, while those from human umbilical vein (HUVEC) are polygonal [Fuchs et al. 2001;

Lang et al. 2003].

26

Page 42: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Furthermore, endothelial cells derived from arteries are different than those isolated from

veins (Figure 2a) [Aird 2007; Lang et al. 2003]. In arteries, endothelial cells are long and

narrow, aligned in the direction of blood flow and form a continuous endothelium with many

tight junctions. Endothelial cells from veins are shorter and wider, and not aligned in the

direction of blood flow [Aird 2007]. These heterogeneities reflect their difference in

functionality in term of the release of vasoactive substances and their interaction with

leucocytes during normal vessel development and wound healing [Aird 2007; Lang et al.

2003; Otto etal. 2001].

Some unique molecular markers as well as genes, preferentially expressed by endothelial cells,

are listed in Table 2.1. For example, the expression of CD31 (also known as platelet

endothelial cell adhesion molecule-1 or PECAM-1), CD34, and von Willebrand factor (vWF)

as well as Dil-acetylated LDL uptake has been used in many studies to distinguish endothelial

cells from other cell types [Bramfeld et al. Submitted-2010; Hillen et al. 2006; Aird 2007].

Also, cell adhesion molecules (CAM), such as ICAM-1 (intercellular adhesion molecule-1),

VCAM-1 (vascular cell adhesion molecule-1), E- and P-selectins can be used to identify

endothelial cells during wound healing and angiogenesis [Monchaux and Vermette 2010; Otto

et al. 2001; Peters et al. 2008; Kirkpatrick et al. 2003].

Other cell-cell adhesive proteins, such as ESAM (endothelial cell selective adhesion

molecule), VE-cadherin (vascular endothelial cadherin) and N-cadherin, are known to have a

significant in angiogenesis regulation [Dejana 2004; Fuchs et al. 2001; Hillen et al. 2006].

ESAM is a tight junction molecule that is responsible for the regulation of cellular

permeability and for maintaining the polarity of endothelial cells [Dejana 2004; Fuchs et al.

2001]. VE-cadherin is an adherens junction molecule that plays an important role in regulating

cell growth and in the organization of new vessels during angiogenesis [Bazzoni and Dejana

2004; Hillen et al. 2006]. Another member of the cadherin family, N-cadherin is localized on

the basal side of endothelial cells [Hillen et al 2006]. During vessel maturation, N-cadherin is

in contact with pericytes or smooth muscle cells [Bazzoni and Dejana 2004; Dejana 2004].

27

Page 43: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Furthermore, in other angiogenesis studies, the expression of some genes to assess if

endothelial cells are undergoing angiogenesis has been suggested. Some genes that are

preferentially expressed during endothelial cell sprouting, lumen formation and capillary

network establishment are HESR-1, notch 1, notch 4 and delta 4 [Nakatsu et al. 2003a;

Sainson et al. 2005; Henderson et al. 2001; Taylor et al. 2002].

2.5.1 Vasculogenesis and angiogenesis

Recent studies on blood microvessels have provided essential information to develop

strategies allowing neovascularization. Microvessels can develop through two processes:

vasculogenesis and angiogenesis. Both can be potentially applied to vascularize tissue

constructs. Although in some papers both terms are often used to mean the same thing and are

simply referred to as angiogenesis, the specific role of the micro-environmental factors and the

overall mechanisms are different [Francis et al. 2008].

Vasculogenesis refers to the process of differentiation of endothelial progenitor cells (i.e.,

mesodermal, mesenchymal or bone marrow cells) to form new blood vessels. For example, in

dermal tissues, vasculogenesis occurs in three main steps: 1) differentiation of mesodermal

cells into angioblasts or hemangioblasts; 2) differentiation of angioblasts or hemangioblasts

into endothelial cells; and 3) the organization of new endothelial cells into a primary capillary

plexus [Moon and West 2008].

Angiogenesis can occur in two different ways: 1) intussusceptive, the longitudinal splitting of

existing vessels, and 2) sprouting angiogenesis, the outgrowth of a new branch from a pre­

existing vessel [Cockerill et al. 1995; Levin et al. 2007; Risau 1997]. Compared to the

intussusceptive angiogenesis that mostly occurs during new organ formation as well as during

tumour development, sprouting angiogenesis is relatively well characterized [Levin et al.

2007]. Here, I will focus on sprouting angiogenesis. Sprouting angiogenesis refers to the

formation of new capillaries from pre-existing blood vessels, which is mainly initiated by a

sprouting process [Cockerill et al. 1995; Risau 1997].

28

Page 44: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Angiogenesis involves four different stages: 1) endothelial cells interact with their ECM and

the underlying basement membrane; 2) they proliferate, migrate and communicate with each

others; 3) they form cell-cell connections and tube-like structures; and 4) tube sprouting and

remodelling occur to form microvessels containing multi-cellular lumen [Sukmana and

Vermette 2010a; Risau 1997].

Our knowledge of angiogenesis as well as the availability of numerous models was pioneered

about 30 years ago by Folkman and Haudenschild (1980) when they demonstrated that new

capillary blood vessels form in tumor progression. They observed that capillary tube formation

was initiated from a vacuole structure within the endothelial cells that subsequently develop

multi-cellular capillary lumen [Folkman and Haudenschild 1980]. More recently, Kucera et al.

(2007) have proposed three models explaining how tube-like structures developed during the

vascularization process: 1) Cell death and phagocytosis. 2) Wrapping of the spaces around the

extracellular matrix, and 3) vacuole formation from the coalescence of intracellular vacuoles.

Below, I will briefly discuss these three models [Kucera et al. 2007]. The models are presented

on the Figure 2.2.

2.5.1.1 Model 1: cell death and phagocytosis

Some in vitro as well as in vivo studies of angiogenesis report that lumen formation was

associated with apoptotic and cell death events [Fierlbeck et al. 2003; Meyer et al. 1997]. This

model proposed the following steps in vascular lumen formation: endothelial cells organize

themselves into a multi-cellular cord and cells in the middle of the cord become apoptotic and

die. Finally, endothelial cells at the edge phagocytise the apoptotic cells, subsequently forming

a multi-cellular vessel containing lumen (see Figure la). According to this model, dead cells

define the lumen of the tube-like structures. However, this model fails to explain how the

tube-like structures continue their formation into a continuous vessel [Kucera et al. 2007].

2.5.1.2 Model 2: wrapping spaces around ECM

This model mainly suggests that most capillary are initiated by the elongation of endothelial

cells to open multi-cellular structures. Subsequently, vascular tube can form when the

elongated cell sheets are closed, even without vacuole formation (see Figure lb) [Kucera et al.

29

Page 45: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2007]. This concept was initiated by Hirakow and Hiruma (1983), when they studied the

development of vascular lumen made of endothelial cells without the evidence of vacuole-like

structures. In a recent study, Parker et al. (2004) have shown that the vascular development

was initiated by the proliferation and migration of endothelial cells to form a cord-like

structure, and vascular cords then undergo tubulogenesis to form vessels containing lumen.

However, this model does not explain the polarity of endothelial cells during the development

of capillary structures [Kucera et al. 2007].

' Lumen %'

Phagocytosis" and exocytosis

Lumen levelopmen

b

Elongation * Closure and IS lumen formation

m Membrane *

invagination

fff Vacuole

Vacuole formation

Lumen

Vacuole fusion & lumen development

"f^ Extracellular matrix

^,r Endothelial cell

Apoptoticcell

Figure 2.2: Models of the development of tube-like structures during vasculogenesis. Adapted

from Kucera et al. (2007).

30

Page 46: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2.5.1.3 Model 3: vacuole formation from the coalescence of intracellular vacuoles

The idea behind this model was proposed by Folkman and Haudenschild (1980) when

explaining angiogenesis and lumen formation within 3D constructs. This model involves

several steps: formation either of single or multiple vacuole(s) inside the endothelial cells and

vacuoles subsequently fuse with each others. Finally, a continuous multi-cellular lumen will

form when two or more cells with their vacuoles adhere to each others, therefore fusing (see

Figure lc) [Folkman and Haudenschild 1980; Davis et al. 2000].

Davis et al. (2000) have reported that endothelial cell lumens were forming in three-

dimensional collagen and fibrin matrices. The authors reported that this process was controlled

by the formation and coalescence of intracellular vacuoles and in the absence of cellular

junctions [Davis et al. 2000]. Furthermore, the expression of cytoskeletal regulators

controlling various cellular functions of endothelial cells, such as Cdc42 and Racl GTPases,

' were involved in morphogenesis and vascularization processes [Connolly et al. 2002; Bayless

and Davis 2002].

Even though this model can explain the role of vacuoles in capillary tube structure and lumen

formation, some questions remain, such as how endothelial cells compensate their basal side

during the vacuole coalescence and vacuole fusion, and how the apical-basal polarity of

endothelial cells can be established during tube stabilization [Kucera et al. 2007; Yancopoulos

2000]. Since none of these three models seem to include all the cellular events observed

during vascular tube development, it can thus be hypothesized that all these three models can

be combined, in some way, to explain observations made in angiogenesis and vasculogenesis

assays [Darland and D'Amore 2001; Hubbel 2003; Lutolf and Hubbel 2005].

2.5.2 Promoting angiogenesis in three-dimensional constructs

During vasculogenesis and angiogenesis development, cell-cell as well as cell-ECM

interactions are complex [Darland and D'Amore 2001; Lutolf and Hubbel 2005]. While the

roles of some individual factors during neovascularization have been investigated, the

optimum combination between cell and their ECM can significantly vary from one material to

31

Page 47: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

another [Hubbel 2003]. Therefore, some strategies have been proposed to promote and direct

angiogenesis in 3D environments.

Endothelial cells have been cultured in various matrices that have been pre-coated or not with

adhesive matrix proteins or cell binding peptides. Surface chemistry and the immobilization

mode can affect protein conformation, orientation and anchorage strength, thus influencing

cell behaviour [Hubbel 2003]. Formation of capillary-like structures made of endothelial cells

depends on the particular biological environment that will direct cell-cell as well as cell-

biomaterial adhesion strength, which relies on biochemical and mechanical signals [Kannan et

al. 2005]. To date, there are three major strategies that have been used to study how

biomaterials properties affect angiogenesis formation: 1) Micro-patterning, 2) Surface

modification, and 3) extracellular matrix modifications [Kannan et al. 2005; Moon and West

2008].

2.5.2.1 Micro-patterning

Photolithography, micro-moulding, and micro-printing are some examples of techniques used

in micro-patterning. Among them, laser micro-contact printing is the most frequently studied

[Hahn et al. 2006; Kannan et al. 2005]. It is a process in which biological molecules are

printed directly onto a scaffold surface, and it has been broadly utilized by tissue engineering

scientists. For example, a poly(ethylene glycol)-diacrylate (PEGDA) hydrogel, known to resist

protein adsorption, has been micro-printed with the cell adhesive ligand, Arg-Gly-Asp-Ser

(RGDS) in different concentrations and using different patterns. Endothelial cells were

cultured on these RGDS patterns and enhanced tube-like structure formation was observed on

50u,m-wide stripes [Moon and West 2008].

In another study, micro-contact printing was used to form various patterns of fibronectin

molecules on gold [Moon and West 2008]. Endothelial cells cultured on these fibronectin

patterns formed tubular structures on lOum-wide lines of fibronectin [Moldovan and Ferrari

2002]. In a different study, when coated with gelatin, 20u.m lines promoted endothelial cells to

undergo capillary morphogenesis [Medalie et al. 1996]. However, applying this technique to

3D scaffolds is problematic [Kannan et al. 2005].

32

Page 48: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2.5.2.2 Surface modification

Proteins such as fibronectin, gelatin, collagens, and fibrinogen, only to name a few, have been

used to coat synthetic polymers to increase their surface bioactivity [Ye et al. 2000a]. For

example, collagen coating on the surface of electrospun poly(L-lactic acid)-co-poly(e-

caprolactone) scaffold increased endothelial cell adhesion and spreading [He et al. 2005; Pham

et al. 2006]. Collagen type I has been known to increase the number of tube-like structures

made of endothelial cells, thus supporting angiogenesis and vasculogenesis in in vitro

experiments [Davis and Sanger 2005]. Furthermore, as previously described, the proteolytic

cleavage of collagen type IV provides an important binding site for endothelial cells

undergoing angiogenesis in vitro [Xu et al. 2001].

Gelatin is used by many research groups to pre-coat tissue culture dishes. In tissue

engineering, gelatin was used to coat poly(glycolic acid) (PGA) scaffold for the controlled

release of some angiogenic growth factors [Dcada and Tabata 1998; Tabata et al. 1994; Tabata

and Ikada 1999]. When PGA scaffolds covered by bFGF- and VEGF-containing gelatin

hydrogel were implanted in mice, significant angiogenic effect was observed at the implanted

site [Dcada and Tabata 1998; Tabata 2009]. Further clinical test to treat diabetic foot ulcer

using gelatin matrix to release bFGF confirmed that the matrix allowed better wound healing

[Tabata 2009].

Fibronectin has been investigated in many studies and found to enhance endothelial cell

adhesion and spreading [Kannan et al. 2005; Monchaux and Vermette 2010]. Fibronectin also

supports the formation of focal adhesion and induces the organization of actin filaments into

stress fibre via the interaction with its main receptor i.e., a5|31 and avP3 [Burmeister et al.

1996; van Wachem et al. 1997]. Furthermore, the lack of fibronectin during vascular

development caused an abnormality in the vascular formation in mice [Burmeister et al. 1996;

George et al. 1997].

Another protein, fibrinogen, has been incorporated in poly(ethylene glycol) (PEG) hydrogels

and it found to increase the bioactivity of the scaffold by enhancing endothelial cell and SMC

adhesion [Almany and Seliktar 2005] as well as mesenchymal stem cell adhesion [Zhang et al.

33

Page 49: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

2006]. Furthermore, fibrinogen has many binding sites for endothelial cells. Also, fibrin gel

binds many growth factors and bioactive cloth components [Herrick et al. 1999; Weisel 2005].

Therefore, the use of fibrin(ogen) to modify biomaterials surfaces and to encapsulate cells in

tissue engineering applications is of importance [Janmey et al. 2009; Tawil 2006].

In other strategies, polymer surfaces can be pre-coated with cells to increase the bioactivity of

the scaffold. For example, expanded poly(tetrafluoroethylene) (ePTFE) was pre-coated with

bladder carcinoma cells before being implanted in mice. This strategy stimulated angiogenesis

and neovascularization in ePTFE vascular grafts [Kidd et al. 2002]. In a more recent study,

pre-coated PET fibres with HUVEC allowed the guidance of the angiogenesis process and

subsequent micro-vascularization in fibrin [Sukmana and Vermette 2010b]. No microvessel

structures were observed when uncovered fibres were utilized (see Chapter 4 of this thesis).

2.5.2.3 ECM modification

The interaction between cells and their matrix is very complex, since there are many proteins

and soluble molecules (including growth factors) present in the ECM [Davis et al. 2002; Davis

and Sanger 2005; Ingber and Folkman 1989]. These molecules interact with cells through

different cell binding domains. In blood vessel development, there are approximately 20

angiogenic growth factors, and among them vascular endothelial growth factor (VEGF), basic

fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF) and angiopoietin-1

(Agp-1) are the most frequently studied [Nakatsu et al. 2003b].

In in vitro angiogenesis assays, growth factors can be added to the culture media and/or

immobilized in the ECM. These strategies have been found to have a significant effect on the

development of tube-like structures and on microvessel maturation [Fourier and Doillon 2007;

Zisch et al. 2003]. For example, endothelial cells cultured in type I collagen gels with culture

media supplemented with fibroblast growth factor-2 (FGF-2), vascular endothelial growth

factor (VEGF), and phorbol ester formed tubes containing lumens, which have similar

structure to microvessels found in vivo [Davis and Camarillo 1996]. In the absence of growth

factors and phorbol ester, the vascular tube structures did not form [Davis and Camarillo 1996;

Kannan 2005]. Other in vitro angiogenesis studies showed that growth factors added to culture

34

Page 50: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

media can promote sprouting, lumen formation and better vessel stability [Chen et al. 2009;

Nakatsu et al. 2003b; Pepper 1997; Uemura et al. 2002].

Growth factors are often bound to the ECM and can be released upon interaction with cells

[Moon et al. 2009; Moon and West 2008]. In some studies, growth factors were immobilized

to the ECM that was used to carry out the culture [Hubbel 2007; Lutolf and Hubbel 2005]. In

this approach, growth factors can be released, as encountered in vivo. For example, VEGF was

covalently immobilized into type I collagen gel [Koch et al. 2006]. Using the chicken

chorioallantoic membrane (CAM) assay, this collagen matrix was found to induce capillary

formation and tissue ingrowth [Koch et al. 2006]. Also, basic fibroblast growth factor (bFGF)

was immobilized into poly(ethylene glycol) (PEG) hydrogels to guide cell alignment and

migration [DeLong et al. 2005; Koch et al. 2006; Moon et al. 2009].

As many cellular processes observed during vasculogenesis require numerous signalling

pathways and growth factors, recent research efforts have been focusing on the sequential

delivery of multiple growth factors [Mooney et al. 1997]. For example, the sequential delivery

of VEGF and PDGF-BB (platelet-derived growth factor-BB) in a controlled-release polymer

device made of poly(lactic glycolic) (PLG) (85:15 lactide:glycolide) induced a mature

vascular network covered by smooth muscle cells [Richardson et al. 2001; Sumpio et al.

1998]. Other results also concluded that one growth factor alone is not sufficient to create

mature and stable vasculature [Blau et al. 2001; Mooney et al. 1997].

2.5.3 Co-culture systems and microvessel maturation

Achieving microvessel maturation and stabilization during blood microvessel development

represents another challenge following vascular tube formation by endothelial cells. This is

often referred to as angiogenesis remodelling [Hubbel 2003; Yancopoulos 2000].

Angiogenesis remodelling is the process by which primary vascular tubes or immature vessels

are modified to form an interconnecting branching network, yielding to microvessel structure

stabilization [Ennett and Mooney 2002; Hubbel 2003]. During this phase, tube-like structures

made of endothelial cells and containing lumens recruit other supporting cells, such as

35

Page 51: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

pericytes and smooth muscle cells (SMC), to form a close wall and mature blood microvessels

[Hubbel 2003; Jain 2003].

Furthermore, in the vascular tree, endothelial cells are lining on the inner layer of the vessel

wall - this is the tunica intima - and are supported by other cell types [Jain 2003; Korff et al.

2001]. For example, immature vessels consist of tube structures made of endothelial cells.

Capillaries consist of tubes made of endothelial cells covered by pericytes and a basement

membrane. In arterioles and venules, vascular SMC cover large area and are closely associated

with the endothelium basement membrane. Finally, as discussed in a previous section, the

structure of large veins and arteries consists of three distinct layers: tunica intima, tunica

media and tunica adventia [Carmeliet and Conway 2001; Jain 2003]. The reader is referred to

Figure 3 for schematic illustrations of the different vessel structures.

Figure 2.3: Blood vessel structure and development. Adapted from Jain (2003).

36

Page 52: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Therefore, co-culture systems seem to be appealing to recreate the real microvessel

environment and to achieve more mature microvessels. For example, smaller number of

vessel-like structures containing lumens was found when endothelial cells were cultured alone

in PLLA-PLGA (50:50) scaffolds, compared to samples co-cultured with fibroblasts

[Levenberg et al. 2005]. Vascularized engineered skeletal muscle tissue constructs have been

successfully produced by Levenberg et al. using a multiculture system of myoblasts

(progenitor cells of the muscle cells), fibroblasts and endothelial cells into this PLLA-PLGA

scaffold [Levenberg et al. 2005].

2.6 Bioreactors to support vascularization

Many tissue engineering scientists believe that the next generation of functional tissue

replacements truly needs the use of bioreactors, in which the culture conditions (e.g., pH,

temperature, O2 concentration, pressure, pulsation, nutrient transfer and waste removal, etc.)

can be adjusted and studied to find the optimal mechanical, chemical, and biological stimuli

for a given application [Martin et al. 2004]. In addition, bioreactor operations will provide a

rational basis for the structural and functional design of engineered tissues for the use as

model systems to reduce time of innovation, discovery and production in biological and

clinical research [Martin et al. 2004; Martin and Vermette 2005]. Also, the use of bioreactors

should accelerate the development, evaluation, and delivery of engineered tissue products to

patients [Freed et al. 2006].

To date, there are various types of bioreactors that have been designed and tested and these

differ from the applications involved. For example, bioreactors have been used to support the

growth of cartilage [Obradovic et al. 1999; Pazzano et al. 2000; Seidel et al. 2004], bone

[Ignatius et al. 2005; Yu et al. 2004], cardiac [Carrier et al. 2002; Radisic et al. 2004],

ligament [Alltman et al. 2002; Lee et al. 2005], and heart valve [Hoerstrup et al. 2000;

Schenke-Layland et al. 2003] tissues.

Furthermore, there are some types of bioreactors that can be used in large tissue cultures, and

these include the continuous stirred-tank reactor (CSTR), hollow fiber reactors and the slow

turning lateral vessel (STLV) reactor [Martin and Vermette 2005], only to name a few. CSTR

37

Page 53: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

reactors are designed to mix oxygen and nutrients within the culture medium and to reduce the

boundary layer at the scaffold surface [Belfort 1989]. Hollow fibre reactors consist of a

chamber perfused with semi-permeable fibres filled with culture medium. Often, cells are

located in the extracapillary space of the chamber. In the past years, this type of reactor was

used to produce proteins made by mammalian cells, and to expand mammalian cells in vitro

[Jauregui et al. 1996; Schwarz et al. 1992]. STLV reactor was proposed by NASA (National

Aeronautics and Space Administration) at the Johnson Space Center (USA) for earth-based as

well as space experiments [Schwarz et al. 1992].

In the context of vascularization and blood microvessel development, ideally the bioreactor

should enable to control basic environmental parameters such as the dissolved oxygen

concentration, pH, and temperature, as well as tissue factors including cellular structures and

function [Bilodeau and Mantovani 2006]. For example, the system should allow the transport

of nutrients and oxygen from the reservoir to the cell location within the scaffold. As it has

been long known, the maximum tissue thickness achievable by diffusive transport alone is

approximately 1mm [Martin and Vermette 2005]. The optimization of scaffold material

properties as well as the design of bioreactor chamber is therefore of upmost importance

[Chouinard et al. 2009].

Furthermore, bioreactors can also enable control over the mechanical stimulation for cellular

guidance inside the scaffold in order to improve tissue vascularization. As endothelial cells are

lining the inner surface of blood vessels of the entire capillary and circulatory system, they

experience fluid shear stress and dynamic flow conditions [Aird 2007; Xu et al. 2001].

Therefore, the use of a dynamic culture system can allow some cell mechanical stimulation

prior to the use of the grown tissue construct [Levenberg et al. 2005; Lawrence and Madihally

2008].

Dynamic culture parameters such as pulsation, pressure, and flow rate that mimic in vivo

conditions often result in better cellular organization as well as mechanical properties of the

tissue constructs when compared to constructs cultured in static environment [Sodian et al.

2002; Yao et al. 2005]. To date, several studies of endothelial cell monolayers have revealed

38

Page 54: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

that cell orientation reflects the direction of the flow [Lesman et al. 2010; Li et al. 2005;

Stegemann and Nerem 2003]. Also, in endothelial cells, shear stress increased VEGF [Conklin

et al. 2002] and PDGF-BB expression [Sumpio et al. 1998] compared to those cultured in

static conditions. In addition, pulsatile flow can improve structural organization of SMC and

this is an essential step in the vascularization process [Akhyari et al. 2002; Kofidis et al. 2002;

Niklason et al. 1999].

2.7 Concluding remarks

Over the past years, significant advances in tissue engineering research have provided hope for

the commercial availability of human bioartificial materials. More simple engineered tissue

substitutes (e.g., skin, bone, and cartilage) have been successfully applied and implanted,

while for more complex and vascularized tissues, many problems still remain before tissue

substitutes can be available. Consequently, the development of biomaterials to engineer tissue

constructs as well as strategies to enhance vascularization inside tissue constructs becomes

important to provide scaffolds with improved oxygen and nutrient transfer as well as waste

removal. This could allow production of thicker tissue substitutes as well as artificial organs.

Tissue engineering research relies on the increasing knowledge of angiogenesis and

vasculogenesis mechanisms occurring during capillary tube formation and blood microvessel

development. Furthermore, to overcome the bottleneck of the complex interplay between

various factors influencing tissue vascularization, the use of bioreactor system is necessary.

39

Page 55: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

References

Agrawal, C. M. and Ray, R. B. (2001) Biodegradable polymeric scaffolds for musculoskeletal

tissue engineering. J.Biomed.Mater.Res. 55, p. 141-50.

Aird, W. C. (2007) Phenotypic heterogeneity of the endothelium: II. Representative vascular

beds. Circ.Res. 100, p. 158-73.

Akhyari, P., et al. (2002) Mechanical stretch regimen enhances the formation of bioengineered

autologous cardiac muscle grafts. Circulation 106, p. 1137-1142.

Allt, G. and Lawrenson, J. G. (2001) Pericytes: cell biology and pathology. Cells Tissues

Organs 169, p. 1-11.

Almany, L. and Seliktar, D. (2005) Biosynthetic hydrogel scaffolds made from fibrinogen and

polyethylene glycol for 3D cell cultures. Biomaterials 26, p. 2467-77.

Altman, G. H., et al. (2002) Advanced bioreactor with controlled application of multi­

dimensional strain for tissue engineering. J.Biomech.Eng. 124, p. 742-49.

Angele, P., et al. (2004) Influence of different collagen species on physico-chemical properties

of crosslinked collagen matrices. Biomaterials 25, p. 2831-41.

Arthur, W. T., et al. (1998) Growth factors reverse the impaired sprouting of microvessels

from aged mice. Microvasc.Res. 55, p. 260-270

Atala, A. (2004) Tissue engineering and regenerative medicine: concepts for clinical

application. Rejuvenation.Res. 7, p. 15-31.

Auerbach, R., et al. (1998) Angiogenesis assays: a critical overview. Clin.Chem. 49, p. 32-40.

Bach, F. H. (1998) Xenotransplantation: problems and prospects. Annu.Rev.Med. 49, p. 301-

10.

Bayless, K. J. and G. E. Davis. (2003) Sphingosine-1 -phosphate markedly induces matrix

metalloproteinase and integrin-dependent human endothelial cell invasion and lumen

formation in three-dimensional collagen and fibrin matrices.

Biochem.Biophys.Res.Commun. 312, p. 903-13.

Bayless, K. J. and Davis, G. E. (2002) The Cdc42 and Racl GTPases are required for capillary

lumen formation in three-dimensional extracellular matrices. J.Cell Sci. 115, p. 1123-36.

Bazzoni, G. and Dejana, E. (2004) Endothelial cell-to-cell junctions: molecular organization

and role in vascular homeostasis. Physiol.Rev. 84, p. 869-901.

40

Page 56: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Becker et al. (2009) Extracellular Matrix. In The World of the cell, chapter 11. 7 edition.

Pearson/Benjamin Cummings, San Francisco. 791 p.

Belfort, G. (1989) Membranes and bioreactors: a technical challenge in biotechnology.

Biotechnol.Bioeng. 33, p. 1047-66.

Bilodeau, K. and Mantovani, D. (2006) Bioreactors for tissue engineering: focus on

mechanical constraints. A comparative review. Tissue Eng. 12, p. 2367-83.

Black, A. F., et al. (1999) A novel approach for studying angiogenesis: a human skin

equivalent with a capillary-like network. Cell Biol.Toxicol. 15, p. 81-90.

Blau, H. M., Brazelton, T. R. and Weimann, J. M. (2001) The evolving concept of a stem cell:

entity or function? Cell 105, p. 829-41.

Bramfeld, H., et al. (2010) Scaffold vascularisation: a challenge for three-dimensional tissue

engineering. Curr.Med.Chem. Submitted.

Bramfeldt, H., Sarazin, P. and Vermette, P. (2007) Characterization, degradation, and

mechanical strength of poly(D,L-lactide-co-epsilon-caprolactone)-poly(ethylene glycol)-

poly(D,L-lactide-co-epsilon-caprolactone). J.Biomed.Mater.ResA 83, p. 503-11.

Brekke, J. H., et al. (2006) Hyaluronan as a biomaterials. In Guelcher, S.A. and Hollinger, J.O.

(editors) An introduction to Biomaterials 2n edition. CRC Press, Boca Raton, p. 219-

248.

Burmeister, J. S., et al. (1996) Effect of fibroriectin amount and conformation on the strength

of endothelial cell adhesion to HEMA/EMA copolymers. J.Biomed.Mater.Res. 30, p. 13-

22.

Carmeliet, P. (2005) Angiogenesis in life, disease and medicine. Nature 438, p. 932-936.

Carmeliet, P. and Conway, E. M. (2001) Growing better blood vessels. Nat.Biotechnol. 19, p.

1019-1020.

Carrier, R. L., et al. (2002) Perfusion improves tissue architecture of engineered cardiac

muscle. Tissue Eng. 8, p. 175-88.

Catelas, I., et al. (2006) Human mesenchymal stem cell proliferation and osteogenic

differentiation in fibrin gels in vitro. Tissue Eng. 12, p. 2385-96.

Chaikof, E. L., et al. (2002) Biomaterials and scaffolds in reparative medicine.

Ann.N.Y.Acad.Sci. 961, p. 96-105.

41

Page 57: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Chang, C. and Werb, Z. (2001) The many faces of metalloproteases: cell growth, invasion,

angiogenesis and metastasis. Trends Cell Biol. 11, p. S37-S43.

Chang, Y. K., Nair, K. and Sun, W. (2010) Three dimensional multi-scale modelling and

analysis of cell damage in cell-encapsulated alginate constructs. J.Biomech. In press.

Chen, Z., et al. (2009) In vitro angiogenesis by human umbilical vein cells (HUVEC) induced

by three-dimensional co-culture with blioblastoma cells. J. Neurooncol. 92, p. 121-8.

Chen, C. S., Yannas, I. V. and Spector, M. (1995) Pore strain behaviour of collagen-

glycosaminoglycan analogues of extracellular matrix. Biomaterials 16, p. lll-%?>.

Cheresh, D. A., et al. (1989) Recognition of distinct adhesive sites on fibrinogen by related

integrins on platelets and endothelial cells. Cell 58, p. 945-53.

Chiu, Y. C , et al. (2009) Materials for engineering vascularized adipose tissue. J.Tissue

Viability. In press.

Chouinard, J. A., et al. (2009) Design and validation of a pulsatile perfusion bioreactor for 3D

high cell density cultures. Biotechnol.Bioeng. 104, p. 1215-23.

Clowes, A.W., et al, (1986) Mechanisms of arterial graft failure. II. Chronic endothelial and

smooth muscle cell proliferation in healing polytetrafluoroethylene prostheses

J.Vasc.Surg. 3, p. 877-84.

Cockerill, G. W., Gamble, J. R. and Vadas, M. A. (1995) Angiogenesis: models and

modulators. lnt.Rev.Cytol. 159, p. 113-60.

Conklin, B. S., et al. (2002) Shear stress regulates occludin and VEGF expression in porcine

arterial endothelial cells. J.Surg.Res. 102, p. 13-21.

Connolly, J. O., et al. (2002) Rac regulates endothelial morphogenesis and capillary assembly.

Mol.Biol.Cell 13, p. 2474-85.

Conway, E. M. and Carmeliet, P. (2004) The diversity of endothelial cells: a challenge for

therapeutic angiogenesis. Genome Biol. 5, p. 207.

Cox, S., Cole, M. and Tawil, B. (2004) Behavior of human dermal fibroblasts in three-

dimensional fibrin clots: dependence on fibrinogen and thrombin concentration. Tissue

Eng. 10, p. 942-54.

Darland, D. C. and D'Amore, P. A. (2001) Cell-cell interactions in vascular development.

Curr.Top.Dev.Biol. 52, p. 107-49.

42

Page 58: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Davis, G. E. and Camarillo, C. W. (1996) An alpha 2 beta 1 integrin-dependent pinocytic

mechanism involving intracellular vacuole formation and coalescence regulates capillary

lumen and tube formation in three-dimensional collagen matrix. Exp.Cell Res. 224, p.

39-51.

Davis, G. E. and Senger, D. R. (2005) Endothelial extracellular matrix: biosynthesis,

remodeling, and functions during vascular morphogenesis and neovessel stabilization.

Circ.Res. 97, p. 1093-107.

Davis, G. E., Bayless, K. J. and Mavila, A. (2002) Molecular basis of endothelial cell

morphogenesis in three-dimensional extracellular matrices. Anat.Rec. 268, p. 252-75.

Davis, G. E., Black, S. M. and Bayless, K. J. (2000) Capillary morphogenesis during human

endothelial cell invasion of three-dimensional collagen matrices. InVitroCell

Dev.Biol.Anim. 36, p. 513-19.

Dejana, E., et al. (1987) Fibrinogen induces adhesion, spreading and microfilament

organization of human endothelial cell in vitro. J Cell Biol. 105, p. 1403-11.

Dejana, E. (2004) Endothelial cell-cell junctions: happy together. Nat.Rev.Mol.Cell Biol. 5, p.

261-70.

DeLong, S. A., Moon, J. J. and West, J. L. (2005) Covalently immobilized gradients of bFGF

on hydrogel scaffolds for directed cell migration. Biomaterials 26, p. 227-34.

Deroanne, C. F., Lapiere, C. M. and Nusgens, B. V. (2001) In vitro tubulogenesis of

endothelial cells by relaxation of the coupling extracellular matrix-cytoskeleton.

Cardiovasc.Res. 49, p. 647-58.

Discher, D. E., Janmey, P. and Wang, Y. L. (2005) Tissue cells feel and respond to the

stiffness of their substrate. Science 310, p. 1139-43.

Draget, K. I., Skjak-Braek, G. and Smidsrod, O. (1997) Alginate based new materials.

Int.J.Biol.Macromol. 21, p. 47-55.

Drury, J. L. and Mooney, D. J. (2003) Hydrogels for tissue engineering: scaffold design

variables and applications. Biomaterials 24, p. 4337-51.

Duband, J. L., et al. (1993) Calponin and SM 22 as differentiation markers of smooth muscle:

spatiotemporal distribution during avian embryonic development. Differentiation 55, p.

1-11.

43

Page 59: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Duong, H., Wu, B. and Tawil, (2009) Modulation of 3D fibrin matrix stiffness by intrinsic

fibrinogen-thrombin compositions and by extrinsic cellular activity. Tissue Eng. Part A

15, p. 1865-76.

Ennett, A. B. and Mooney, D. J. (2002) Tissue engineering strategies for in vivo

neovascularisation. Expert.Opin.Biol.Ther. 2, p 805-18.

Ferguson, W. D., Collins, L. M. and Smith, D. W. (2003) Psychophysical threshold variability

in cochlear implant subjects. Hear.Res. 180, p. 101-13.

Ferrenq, I., Tranqui, L., Vailhe, B, Gumery, P.Y., Tracqui, P. (1997) Modelling biological gel

contraction by cells: mechanocellular formulation and cell traction force quantification.

Acta Biotheor. 45, p. 267-93.

Fierlbeck, W., et al. (2003) Endothelial cell apoptosis during glomerular capillary lumen

formation in vivo. J.Am.Soc.Nephrol. 14, p. 1349-54.

Folkman, J. and Haudenschild C. (1980) Angiogenesis in vitro. Nature 288, p. 551-56.

Fourier, N. and Doillon, C.J. (2007) In vitro effects of extracellular matrix and growth factors

on endothelial cell migration and vessel formation. Cells and Materials 4, p. 399-408.

Francis, M. E., Uriel, S. and Brey, E. M. (2008) Endothelial cell-matrix interactions in

neovascularization. Tissue Eng Part B Rev. 14, p. 19-32.

Freed, L. E., et al. (2006) Advanced tools for tissue engineering: scaffolds, bioreactors, and

signaling. Tissue Eng 12, p. 3285-305.

Fuchs, J. R., Nasseri, B. A. and Vacanti. J. P. (2001) Tissue engineering: a 21st century

solution to surgical reconstruction. Ann.Thorac.Surg. 72, p 577-91.

Garcia, A.J., et al. (2005) Tissue Engineering Scaffold. In Fisher J.P. et al. (editors.) Tissue

Engineering 2nd edition. Springer, New York, p. 10-21.

Geiger, B., et al. (2001) Transmembrane crosstalk between the extracellular matrix-

cytoskeleton crosstalk. Nat.Rev.Mol.Cell Biol. 2, p. 793-805.

George, E. L., Baldwin, H. S. and Hynes, R. O. (1997) Fibronectins are essential for heart and

blood vessel morphogenesis but are dispensable for initial specification of precursor

cells, fi/ood 90, p. 3073-81.

Gimble, J. and Guilak, F. (2003) Adipose-derived adult stem cells: isolation, characterization,

and differentiation potential. Cytotherapy 5, p. 362-369.

44

Page 60: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Godbey, W. T. and Atala, A. (2002) In vitro systems for tissue engineering. Ann.N.YAcad.Sci.

961, p. 10-26.

Grizzi, I., et al. (1995) Hydrolytic degradation of devices based on poly(DL-lactic acid) size-

dependence. Biomaterials 16, p. 305-11.

Gullberg, D. E. and Lundgren-Akerlund, E. (2002) Collagen-binding I domain integrins-what

do they do? Prog.Histochem.Cytochem. 37, p. 3-54.

Haarer, J.C., and Dee, K. C. (2006) Proteins and amino acid-derived polymers. In Guelcher

S.A. and Hollinger, J.O. (editors) An Introduction to Biomaterials 2nd edition. CRC

Press, Boca Raton, p. 121-138.

Hahn, M. S., et al. (2006) Photolithographic patterning of polyethylene glycol hydrogels.

Biomaterials 27, p. 2519-24.

Harding, S. I., et al. (2002) Engineering and cell attachment properties of human fibronectin-

fibrinogen scaffolds for use in tissue engineered blood vessels. Bioprocess.Biosyst.Eng.

25, p. 53-59.

He, W., et al. (2005) Fabrication and endothelialization of collagen-blended biodegradable

polymer nanofibers: potential vascular graft for blood vessel tissue engineering. Tissue

Eng. 11, p. 1574-88.

Henderson, A. M., et al. (2001) The basic helix-loop-helix transcription factor HESR1

regulates endothelial cell tube formation. J.Biol.Chem. 276, p. 6169-76.

Herrick, S., et al. (1999) Fibrinogen. IntJ.Biochem.Cell Biol. 31, p. 741-46.

Hillen, F., et al. (2006) Angiogenesis assay, chapter 1, p. 4-38.

Hirakow, R. and Hiruma, T. (1983) TEM-studies on development and canalization of the

dorsal aorta in the chick embryo. Anat.Embryol.(Berl) 166, p. 307-15.

Hoerstrup, S. P., et al. (2000) New pulsatile bioreactor for in vitro formation of tissue

engineered heart valves. Tissue Eng. 6, p. 75-79.

Howe, A., et al. (1998) Integrin signaling and cell growth control. Curr.Opin.Cell Biol. 10, p.

220-223.

Huang, Y., et al. (2005) In vitro characterization of chitosan-gelatin scaffolds for tissue

engineering. Biomaterials 26, p. 7616-27.

Hubbel, J. A. (2003) Tissue and cell engineering. Curr.Opin.Biotechnol. 14, p. 517-19.

45

Page 61: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Hubbel, J.A. (2007) Matrix-bound growth factors in tissue repair. Swiss.Med. Wkly. 137, p.

72S-76S.

Hungerford, J. E. and Little, C. D. (1999) Developmental biology of the vascular smooth

muscle cell: building a multilayered vessel wall. J.Vasc.Res. 36, p. 2-27.

Hutchings, H., Ortega, N. and Plouet, J. (2003) Extracellular matrix-bound vascular

endothelial growth factor promotes endothelial cell adhesion, migration, and survival

through integrin ligation. FASEB J 17, p. 1520-22.

Hutmacher, D. W. (2001) Scaffold design and fabrication technologies for engineering tissues-

state of the art and future perspectives. J.Biomater.Sci.Polym.Ed. 12, p. 107-24.

Huttenlocher, A., Sandborg, R. R. and Horwitz, A. F. (1995) Adhesion in cell migration.

Curr.Opin.Cell Biol. 7, p. 697-706.

Ignatius, A., et al. (2005) Osseointegration of alumina with a bioactive coating under load-

bearing and unloaded conditions. Biomaterials 26, p. 2325-32.

Ikada, Y. and Tabata, Y. (1998) Protein release from gelatin matrices. Adv.Drug Deliv.Rev.

31, p. 287-301.

Ingber, D.E., et al. (1995) Cell shape, cytoskeletal mechanics and cell cycle control in

angiogenesis. J. Biomechanics 28, p. 1471-84.

Ingber, D. E. and Folkman, J. (1989) Mechanochemical switching between growth and

differentiation during fibroblast growth factor-stimulated angiogenesis in vitro: role of

extracellular matrix. J.Cell Biol. 109, p. 317-30.

Isenberg, B. C , Williams, C. and Tranquillo, R. T. (2006) Small-diameter artificial arteries

engineered in vitro. Circ.Res. 98, p. 25-35.

Jain, R. K. (2003) Molecular regulation of vessel maturation. Nat.Med. 9, p. 685-93.

Janmey, P. A., Winer, J. P. and Weisel, J. W. (2009) Fibrin gels and their clinical and

bioengineering applications. J.R.Soc.Interface. 6, p. 1-10.

Jauregui, H. O., Chowdhury, N. R. and Chowdhury, J. R. (1996) Use of mammalian liver cells

for artificial liver support. Cell Transplant. 5, p. 353-67.

Jung, Y. D., et al. (2002) The role of the microenvironment and intercellular cross-talk in

tumor angiogenesis. Semin. Cancer Biol. 12, p. 105-12.

Kannan, R. Y., et al. (2005) The roles of tissue engineering and vascularisation in the

development of micro-vascular networks: a review. Biomaterials 26, p. 1857-75.

46

Page 62: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Kidd, K. R., et al. (2002) A comparative evaluation of the tissue responses associated with

polymeric implants in the rat and mouse. J.Biomed.Mater.Res. 59, p. 682-89.

Kikuchi, A., et al. (1997) Linear chronic cutaneous graft-versus-host disease.

J.Am.Acad.Dermatol. 37, p. 1004-06.

Kirkpatrick, C. J., et al. (2003) Experimental approaches to study vascularization in tissue

engineering and biomaterial applications. J.Mater.Sci.Mater.Med. 14, p. 677-81.

Ko, H. C , Milthorpe, B. K. and McFarland, C. D. (2007) Engineering thick tissues-the

vascularisation problem. Eur.Cell Mater. 14, p. 1-18.

Koch, S., et al. (2006) Enhancing angiogenesis in collagen matrices by covalent incorporation

of VEGF. J.Mater.Sci.Mater.Med. 17, p. 735-41.

Kofidis, T., et al. (2002) In vitro engineering of heart muscle: artificial myocardial tissue.

J.Thorac.Cardiovasc.Surg. 124, p. 63-69.

Koh, C. J., et al. (2009) Parthenogenesis-derived multipotent stem cells adapted for tissue

engineering applications. Methods 47, p. 90-97.

Korff, T., et al. (2001) Blood vessel maturation in a 3-dimensional spheroidal coculture

model: direct contact with smooth muscle cells regulates endothelial cell quiescence and

abrogates VEGF responsiveness. FASEB J. 15, p. 447-57.

Kucera T, Eglinger J, Strilic B, Lammert E. (2007) Vascular lumen formation from a cell

biological perspective. In: Chadwick, D.J. and Goode, J. (editors) Vascular

Development. West Sussex: John Willey & Sons Ltd. p. 46-56.

Lang, I., et al. (2003) Heterogeneity of microvascular endothelial cells isolated from human

term placenta and macrovascular umbilical vein endothelial cells. Eur. J.Cell Biol. 82, p.

163-73.

Langer, R. and Vacanti, J. P. (2003) Tissue engineering. Science 260, p. 920-926.

Langer, R., et al. (1995) Tissue engineering: biomedical applications. Tissue Eng. 1, p. 151-

161.

Lanza, R. P. and Chick, W. L. (1997) Transplantation of encapsulated cells and tissues.

Surgery 121, p. 1-9.

Lanza, R.P., Langer, R.S. and Vacanti, J. (2000) In Principles of Tissue Engineering, 2nd ed.

Academic Press, San Diego.

47

Page 63: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Larson III, J.W. and Chu, C.R. (2006) Tissue engineering of articular cartilage. In Guelcher

S.A. and Hollinger, J.O. (editors) An Introduction to Biomaterials 2n edition. CRC

Press, Boca Raton, p. 525-536.

Lawrence, B. J. and Madihally, S. V. (2008) Cell colonization in degradable 3D porous

matrices. Cell Adh.Migr. 2, p. 9-16.

Lee, C. H., et al. (2005) Differential pretensions of a flexor tendon graft for anterior cruciate

ligament reconstruction: a biomechanical comparison in a porcine knee model.

Arthroscopy 21, p. 540-46.

Lee, J. W., et al. (2008) Fabrication and characteristic analysis of a poly(propylene fumarate)

scaffold using micro-stereolithography technology. J.Biomed.Mater.Res.B

Appl.Biomater. 87, p. 1-9.

Lesley, J., et al. (2000) Hyaluronan binding by cell surface CD44. J.Biol.Chem. 275, p. 26967-

75.

Lesman, A., Blinder, Y. and Y. Levenberg, Y. (2010) Modeling of flow-induced shear stress

applied on 3D cellular scaffolds: Implications for vascular tissue engineering.

Biotechnol.Bioeng. 105. p. 645-54.

Levenberg, S. and Langer, R. (2004) Advances in tissue engineering. Curr.Top.Dev.Biol. 61,

p. 113-134.

Levenberg, S., et al. (2005) Engineering vascularized skeletal muscle tissue. Nat.Biotechnol.

23, p. 879-884.

Levin, M., et al. (2007) A model of intussusceptive angiogenesis. Novartis.Found.Symp. 283,

p. 37-42.

Li, S., Huang, N. F. and Hsu, S. (2005) Mechanotransduction in endothelial cell migration.

J.Cell Biochem. 96, p. 1110-26.

Linnes, M. P., Ratner, B. D. and Giachelli, C. M. (2007) A fibrinogen-based precision

microporous scaffold for tissue engineering. Biomaterials 28, p. 5298-306.

Lishko, V. K., Kudryk, B., Yakubenko, V.P., Yee, V.C., Ugarova, T.P. (2002) Regulated

unmasking of the cryptic binding site for integrin alpha M beta 2 in the gamma C-

domain of fibrinogen. Biochemistry. 41, p. 12942-51.

Litvinov, R. I., et al. (2005) Polymerization of fibrin: specificity, strength, and stability of

knob-hole interactions studied at the single-molecule level. Blood 106, p. 2944-51.

48

Page 64: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Lo, C. M., et al. (2000) Cell movement is guided by the rigidity of the substrate. Biophys.J. 79,

p. 144-52.

Lokeshwar, V. B. and Selzer, M. G. (2000) Differences in hyaluronic acid-mediated functions

and signaling in arterial, microvessel, and vein-derived human endothelial cells.

J.Biol.Chem. 275. p. 27641-49.

Lutolf, M. P. and Hubbell, J. A. (2005) Synthetic biomaterials as instructive extracellular

microenvironments for morphogenesis in tissue engineering. Nat.Biotechnol. 23, p. 47-

55.

Ma, L., et al. (2004) Biodegradability and cell-mediated contraction of porous collagen

scaffolds: the effect of lysine as a novel crosslinking bridge. J.Biomed.Mater.Res.A 71,

p. 334-42.

Marshall, A., Barker, T., Sage, E., Hauch, K., Ratner., B. (2004) Pore size controls

angiogenesis in subcutaneously implanted porous matrices. World Biomaterials

Congress 7th. 17-21 May 2004. Sydney, Australia.

Martin, I., Wendt, D. and Heberer, M. (2004) The role of bioreactors in tissue engineering.

Trends Biotechnol. 22, p. 80-86.

Martin, Y. and Vermette, P. (2005) Bioreactors for tissue mass culture: design,

characterization, and recent advances. Biomaterials 26, p. 7481-503.

Medalie, D. A., Tompkins, R. G. and Morgan, J. R. (1996) Evaluation of acellular human

dermis as a dermal analog in a composite skin graft. ASAIO J. 42, p. 455-462.

Meyer, G. T., et al. (1997) Lumen formation during angiogenesis in vitro involves phagocytic

activity, formation and secretion of vacuoles, cell death, and capillary tube remodelling

by different populations of endothelial cells. Anat.Rec. 249, p. 327-40.

Mikos, A. G., et al. (1993) Prevascularization of porous biodegradable polymers.

Biotechnol.Bioeng. 42, p. 716-723.

Miletti-Gonzalez, K. E., et al. (2005) The CD44 receptor interacts with P-glycoprotein to

promote cell migration and invasion in cancer. Cancer Res. 65, p. 6660-67.

Moldovan, N. I. and Ferrari, M. (2002) Prospects for microtechnology and nanotechnology in

bioengineering of replacement microvessels. Arch.Pathol.Lab Med. 126, p. 320-24.

Monchaux, E. and Vermette P. (2010) Effects of surface properties and bioactivation of

biomaterials on endothelial cells. Front Biosci.(Schol.Ed.) 2, p. 239-55.

49

Page 65: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Montano, I., et al. (2009) Formation of human capillaries in vitro: the engineering of

prevascularized matrices. Tissue Eng. Part A 15, p. 1-15.

Montesano R et al. (1986) Basic fibroblast growth factor induces angiogenesis in vitro. Proc.

Nati. Acad. Sci. USA 83, p. 7297-7301.

Moon, J. J. and J. L. West. (2008) Vascularization of engineered tissues: approaches to

promote angiogenesis in biomaterials. Curr.Top.Med.Chem. 8, p. 300-10.

Moon, J. J., et al. (2009) Micropatterning of poly(ethylene glycol) diacrylate hydrogels with

biomolecules to regulate and guide endothelial morphogenesis. Tissue Eng. Part A 15, p.

579-85.

Mooney, D. J. and Mikos, A. G. (1999) Growing new organs. Sci.Am. 280, p. 60-65.

Mooney, D. J., et al. (1997) Long-term engraftment of hepatocytes transplanted on

biodegradable polymer sponges. J.Biomed.Mater.Res. 37, p. 413-20.

Mooney, D. J., et al. (1994) Design and fabrication of biodegradable polymer devices to

engineer tubular tissues. Cell Transplant. 3, p. 203-10.

Mooney, D. J., et al. (1996) Stabilized polyglycolic acid fibre-based tubes for tissue

engineering. Biomaterials 17, p. 115-24.

Nakatsu, M. N., et al. (2003a) 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. 66, p. 102-12.

Nakatsu, M. N., et al. (2003b) VEGF(121) and VEGF(165) regulate blood vessel diameter

through vascular endothelial growth factor receptor 2 in an in vitro angiogenesis model.

Lab.lnvest. 83, p. 1873-85.

Nakatsu, M. N. and Hughes, C. C. W. (2008) An optimized three-dimensional in vitro model

for the analysis of angiogenesis. Meth.in Enzym. 443, p. 65-82.

Nehls, V. and Drenckhahn, D. (1995) A novel, microcarrier-based in vitro assay for rapid and

reliable quantification of three-dimensional cell migration and angiogenesis.

Microvasc.Res. 50, p. 311-22.

Nehls, V. and Herrmann, R. (1996) The configuration of fibrin clots determines capillary

morphogenesis and endothelial cell migration. Microvasc.Res. 51, p. 347-64.

Nerem, R. M. (2006) Tissue engineering: the hope, the hype, and the future. Tissue Eng. 12, p.

1143-1150.

50

Page 66: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Nerem, R.M. (2007) Cell-based therapies: From basic biology to replacement, repair, and

regeneration. Biomaterials 28, p. 5074-77.

Neumann, T., Nicholson, B. S. and Sanders, J. E. (2003) Tissue engineering of perfused

microvessels. Microvasc.Res. 66, p. 59-67.

Niklason, L. E., et al. (2002) Functional arteries grown in vitro. Science 284, p. 489-93.

Nomi, M., Atala, A., Coppi, P.D., and Soker, S. (2002) Principals of neovascularization for

tissue engineering. Mol.Aspects Med. 23, p. 463-83.

Obradovic, B., et al. (1999) Gas exchange is essential for bioreactor cultivation of tissue

engineered cartilage. Biotechnol.Bioeng. 63, p. 197-205.

O'Brien, F. J., et al. (2005) The effect of pore size on cell adhesion in collagen-GAG scaffolds.

Biomaterials 26, p. 433-41.

Orlidge, A. and D'Amore, P. A. (1987) Inhibition of capillary endothelial cell growth by

pericytes and smooth muscle cells. J. Cell Biol. 105, p. 1455-62.

Otto, M., et al. (2001) Differential adhesion of polymorphous neutrophilic granulocytes to

macro- and microvascular endothelial cells under flow conditions. Pathobiology 69, p.

159-71.

Owens, G. K., Kumar, M. S. and Wamhoff, B. R. (2004) Molecular regulation of vascular

smooth muscle cell differentiation in development and disease. Physiol.Rev. 84, p. 767-

801.

Park, Y. D., Tirelli, N. and Hubbell, J. A. (2003) Photopolymerized hyaluronic acid-based

hydrogels and interpenetrating networks. Biomaterials 24, p. 893-900.

Parker, L. H., et al. (2004) The endothelial-cell-derived secreted factor Egfl7 regulates

vascular tube formation. Nature 428, p. 754-58.

Pazzano, D., et al. (2000) Comparison of chondrogensis in static and perfused bioreactor

culture. Biotechnol.Prog. 16, p. 893-96.

Pego, A. P., et al. (2003a) Preparation of degradable porous structures based on 1,3-

trimethylene carbonate and D,L-lactide (co)polymers for heart tissue engineering. Tissue

Eng. 9, p. 981-94.

Pego, A. P., et al. (2003b) In vivo behavior of poly(l,3-trimethylene carbonate) and

copolymers of 1,3-trimethylene carbonate with D,L-lactide or epsilon-caprolactone:

Degradation and tissue response. J.Biomed.Mater.Res.A 67, p. 1044-54.

51

Page 67: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Pelham, R. J. Jr. and Wang, Y. (1997) Cell locomotion and focal adhesions are regulated by

substrate flexibility. Proc.Natl.Acad.Sci.U.S.A 94, p. 13661-65.

Pepper, M. S. (1997) Manipulating angiogenesis. From basic science to the bedside.

Arterioscler.Thromb.Vasc.Biol. 17, p. 605-19.

Peters, K., et al. (2008) Cell type-specific aspects in biocompatibility testing: the intercellular

contact in vitro as an indicator for endothelial cell compatibility. J.Mater.Sci.Mater.Med.

19, p. 1637-44.

Pham, Q. P., Sharma, U. and Mikos, A. G. (2006) Electrospun poly(epsilon-caprolactone)

microfiber and multilayer nanofiber/microfiber scaffolds: characterization of scaffolds

and measurement of cellular infiltration. Biomacromolecules 7, p. 2796-805.

Radisic, M., et al. (2004) Medium perfusion enables engineering of compact and contractile

cardiac tissue. Am. J .Physiol Heart Circ.Physiol 286, p. H507-H516.

Rao, C. M., et al. (1997) Regulation of cellular phosphorylation of hyaluronan binding protein

and its role in the formation of second messenger. Biochim.Biophys.Acta 1336, p. 387-

93.

Rauh, F. and Dornish, M. (2006) Chitosan. In Guelcher, S.A. and Hollinger, J.O. (editors) An

Introduction to Biomaterials 2n edition. CRC Press, Boca Raton, p. 249-259.

Richardson, T. P., et al. (2001) Polymeric system for dual growth factor delivery.

Nat.Biotechnol. 19, p. 1029-34.

Risau, W. (1997) Mechanisms of angiogenesis. Nature 386, p. 671-74.

Rowe, S. L., Lee, S. and Stegemann, J. P. (2007) Influence of thrombin concentration on the

mechanical and morphological properties of cell-seeded fibrin hydrogels. Acta Biomater.

3, p. 59-67.

Sainson, R. C. A., et al. (2005) Cell-autonomous notch signalling regulates endothelial cell

brancing and proliferation during vascular tubulogenesis. FASEB J. 19, p. 1027-1051.

Sakakibara, Y., et al. (2002) Prevascularization with gelatin microspheres containing basic

fibroblast growth factor enhances the benefits of cardiomyocyte transplantation.

J.Thorac.Cardiovasc.Surg. 124, p. 50-56.

Schenke-Layland, K., et al. (2003) Complete dynamic repopulation of decellularized heart

valves by application of defined physical signals-an in vitro study. Cardiovasc.Res. 60,

p. 497-509.

52

Page 68: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Schwarz, R. P., Goodwin, T. J. and Wolf, D. A. (1992) Cell culture for three-dimensional

modeling in rotating-wall vessels: an application of simulated microgravity. J.Tissue

Cult.Methods 14, p. 51-57.

Seidel, J. O., et al. (2004) Long-term culture of tissue engineered cartilage in a perfused

chamber with mechanical stimulation. Biorheology 41, p. 445-58.

Senger, D.R., Claffey, K.P., Benes, J.E., Perruzzi, C.A., Sergiou, A.P., Detmar, M. (1997)

Angiogenesis promoted by vascular endothelial growth factor: regulation through

alphalbetal and alpha2betal integrins. Proc.Natl.Acad.Sci.USA 94, p. 13612-7.

Sipe, J. D. (2002) Tissue engineering and reparative medicine. Ann.N.Y.Acad.Sci. 961, p. 1-9.

Sirois, E., et al. (1998) Endothelial cells exposed to erythrocytes under shear stress: An in

vitro study. Biomaterials 19, p. 1925-34.

Slevin, M., Kumar, S. and Gaffney, J. (2002) Angiogenic oligosaccharides of hyaluronan

induce multiple signaling pathways affecting vascular endothelial cell mitogenic and

wound healing responses. J.Biol.Chem. 277, p. 41046-59.

Smidsrod, O. and Skjak-Braek, G. (1990) Alginate as immobilization matrix for cells. Trends

Biotechnol. 8, p. 71-78.

Sodian, R., et al. (2002) Tissue-engineering bioreactors: a new combined cell-seeding and

perfusion system for vascular tissue engineering. Tissue Eng. 8, p. 863-70.

Soker S, Machado M, Atala A. (2000) Systems for therapeutic angiogenesis in tissue

engineering. World J.Urol. 18, p. 10-8.

Stegemann, et al. (2006) In Ma, X. P., and Elisseeff, J. H. (editors) Scaffolding in Tissue

Engineering, chapter 25. Taylor and Francis, Boca Raton.

Stegemann, J. P. and Nerem, R. M. (2003) Phenotype modulation in vascular tissue

engineering using biochemical and mechanical stimulation. Ann.Biomed.Eng 31, p. 391-

402.

Sugiura, H., et al. (2009) In vivo biological responses and bioresorption of tilapia scale

collagen as a potential biomaterial. J.Biomater.Sci.Polym.Ed 20, p. 1353-68.

Sukmana, I. and Vermette, P. (2010a) Polymer fibers as contact guidance to orient

microvascularization in a 3D environment. J.Biomed.Mater.Res.A 92, p. 1587-97.

53

Page 69: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Sukmana I and Vermette P. (2010b) The effect of co-culture with fibroblast and angiogenic

growth factors on microvascular maturation and multi-cellular lumen formation in

HUVEC-oriented polymer fibre constructs. Biomaterials. In press.

Sumpio, B. E., et al. (1998) Regulation of PDGF-B in endothelial cells exposed to cyclic

strain. Arterioscler.Thromb.Vasc.Biol. 18, p. 349-55.

Sumpio, B. E., Riley, J. T. and Dardik, A. (2002) Cells in focus: endothelial cell.

Int.J.Biochem.Cell Biol. 34, p. 1508-12.

Tabata Y. (2009) Biomaterial technology for tissue engineering applications.

J.Control.Release 31, p. 189-199.

Tabata, Y. and Dcada, Y. (1999) Vascularization effect of basic fibroblast growth factor

released from gelatin hydrogels with different biodegradabilities. Biomaterials 20, p.

2169-2175.

Tabata, Y. (2006) Biosynthetic Scaffold. In Ma, X. P., and Elisseeff, J. H. (editors) Scaffolding

in Tissue Engineering. Taylor and Francis, Boca Raton, p. 46-59.

Tabata, Y., Hijikata, S. and flcada, Y. (1994) Enhanced vascularization and tissue granulation

by basic fibroblast growth factor impregnated in gelatin hydrogels. J.Control.Release

31, p. 189-199.

Tassiopoulos, A. K., et al. (2000) Current concepts in chronic venous ulceration.

Eur.J.Vasc.Endovasc.Surg. 20, p. 227-32.

Tawil, B. (2006) Fibrin. In Guelcher, S.A. and Hollinger, J.O. (editors) An Introduction to

Biomaterials 2nd edition. CRC Press, Boca Raton, p. 105-120.

Taylor, K. L., Henderson, A. M. and Hughes, C. C. (2002) Notch activation during endothelial

cell network formation in vitro targets the basic HLH transcription factor HESR-1 and

downregulates VEGFR-2/KDR expression. Microvasc.Res. 64, p. 372-83.

Toman, P. D., et al. (1999) Production of recombinant human type I procollagen homotrimer

in the mammary gland of transgenic mice. Transgenic Res. 8, p. 415-27.

Tonello, C , et al. (2003) In vitro reconstruction of human dermal equivalent enriched with

endothelial cells. Biomaterials 24, p. 1205-11.

Toole, B. P. (2004) Hyaluronan: from extracellular glue to pericellular cue. Nat.Rev.Cancer 4,

p. 528-39.

54

Page 70: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Uemura, A., et al. (2002) Recombinant angiopoietin-1 restores higher-order architecture of

growing blood vessels in mice in the absence of mural cells. J.Clin.Invest. 110, p. 1619-

28.

Vailhe, B., et al. (1998) The formation of tubular structures by endothelial cells is under the

control of fibrinolysis and mechanical factors. Angiogenesis. 2, p. 331-44.

van Wachem, P. B., et al. (1987) The influence of protein adsorption on interactions of

cultured human endothelial cells with polymers. J.Biomed.Mater.Res. 21, p. 701-18.

Vert, M., Mauduit, J. and Li, S. (1994) Biodegradation of PLA/GA polymers: increasing

complexity. Biomaterials 15, p. 1209-13.

Weisel, J. W. (2005) Fibrinogen and fibrin. Adv.Protein Chem. 70, p. 247-99.

West, D. C , et al. (1985) Angiogenesis induced by degradation products of hyaluronic acid.

Science 228, p. 1324-26.

Xu, J., et al. (2001) Proteolytic exposure of a cryptic site within collagen type IV is required

for angiogenesis and tumor growth in vivo. J. Cell Biol. 154, p. 1069-79.

Yancopoulos, G. D., et al. (2000) Vascular-specific growth factors and blood vessel formation.

Nature 407, p. 242-48.

Yang, S., et al. (2001a) The design of scaffolds for use in tissue engineering. Part I.

Traditional factors. Tissue Eng. 7, p. 679-689.

Yang, S., et al. (2001b) Vascular endothelial cell growth factor-driven endothelial tube

formation is mediated by vascular endothelial cell growth factor receptor-2, a kinase

insert domain-containing receptor. Arterioscler.Thromb.Vasc.Biol. 21, p. 1934-40.

Yannas, I. V., et al. (1989) Synthesis and characterization of a model extracellular matrix that

induces partial regeneration of adult mammalian skin. Proc.Natl.Acad.Sci.U.S.A 86, p.

933-37.

Yao, L., et al. (2005) Fibrin-based tissue-engineered blood vessels: differential effects of

biomaterial and culture parameters on mechanical strength and vascular reactivity.

Tissue Eng. 11, p. 991-1003.

Ye, Q., et al. (2000a) Scaffold precoating with human autologous extracellular matrix for

improved cell attachment in cardiovascular tissue engineering. ASAIO J. 46, p. 730-33.

Ye, Q., et al. (2000b) Tissue engineering in cardiovascular surgery: new approach to develop

completely human autologous tissue. Eur J'.Cat-diothorac.Surg. 17, p. 449-54.

55

Page 71: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Yeung, T., et al. (2005) Effects of substrate stiffness on cell morphology, cytoskeletal

structure, and adhesion. Cell Motil.Cytoskeleton 60, p. 24-34.

Yu, X., et al. (2004) Bioreactor-based bone tissue engineering: the influence of dynamic flow

on osteoblast phenotypic expression and matrix mineralization. Proc.Natl.Acad.Sci.USA

101, p. 11203-08.

Zeltinger, J., et al. (2001) Effect of pore size and void fraction on cellular adhesion,

proliferation, and matrix deposition. Tissue Eng. 7, p. 557-72.

Zhang, Z., Wang, Z., Liu, S., Kodama, M. (2004) Pore size, tissue ingrowth, and

endothelialization of small-diameter microporous polyurethane vascular prostheses.

Biomaterials 25, p. 177-87.

Zhang, G., et al. (2006) A PEGylated fibrin patch for mesenchymal stem cell delivery. Tissue

Eng. 12, p. 9-19.

Zielinski, B. A. and Aebischer, P. (1994) Chitosan as a matrix for mammalian cell

encapsulation. Biomaterials 15, p. 1049-56.

Zisch, A. H., et al. (2003) Cell-demanded release of VEGF from synthetic, biointeractive cell

ingrowth matrices for vascularized tissue growth. FASEB J. 17, p. 2260-62.

Zisch, A. H., Lutolf, M. P. and Hubbell, J. A. (2003) Biopolymeric delivery matrices for

angiogenic growth factors. Cardiovasc.Pathol. 12, p. 295-310.

56

Page 72: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

CHAPTER 3

Polymer Fibres as Contact Guidance to Orient

Microvascularization in a 3D Environment

Irza Sukmana1'2, Patrick Vermette1'2

1. Laboratoire de Bioingenierie et de Biophysique de l'Universite de Sherbrooke, Department

of Chemical Engineering, Universite de Sherbrooke, 2500, blvd de l'Universite, Sherbrooke,

QC, Canada, J1K2R1.

2. Research Centre on Aging, Institut universitaire de geriatrie de Sherbrooke, 1036, rue

Belvedere Sud, Sherbrooke, QC, Canada, J1H 4C4.

Article published in J Biomed Mater Res A 92 (2010), p. 1587-1597.

DOI: 10.1002/jbm.a.32479

http://www3.interscience.wiley.com/iournal/122381455/abstract

57

Page 73: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

3.1 Abstract

We describe an in vitro culture process that uses 100-um diameter poly (ethylene

terephthalate) monofilaments as contact guidance of human umbilical vein endothelial cells

(HUVECs) to orient the development of microvessels in a 3D environment. Untreated fibres,

distanced either by 0.05, 0.1, 0.15, or 0.2 mm, were first covered with HUVECs then,

sandwiched between two layers of fibrin gel containing HUVECs. After 2 and 4 days of

culture, cell connections and microvessels were evaluated. Cell connections formed massively

along and in between adjacent fibres that were distanced by 0.05 and 0.1 mm, whereas with

fibres separated by larger distances, connections were rare. After 4 days of culture, the

optimum fibre-to-fibre distance to form microvessels was 0.1 mm. This study reveals that

polymer fibres embedded in gel can be used as guides to direct the microvascularization

process, with potential applications in cancer and cardiovascular research and tissue

engineering.

3.2 Introduction

One of the recent developments in the field of biotechnology is tissue engineering, which is

the application of engineering principles and medical sciences to develop tissue substitutes

that can be used to replace, repair or regenerate damaged, injured, or missing body parts [1-3].

Although massive progress has been made, some major problems are still left to culture

human tissues. The lack of a sufficient supply of nutrients and oxygen to growing tissues in

vitro, as well as waste removal are two important factors that often lead to the failure of the

culture process or even to that of implants [3,4]. One possible strategy for creating three-

dimensional (3D) engineered tissue substitutes in vitro is to develop a scaffold allowing the

guidance of cells in order to promote microvascularization in directional fashion.

Pre-vascularization of tissue-scaffold appears to be the most favorable and efficient approach

to address the problem of tissue survival due to a lack of oxygen supply. Pre-vascularization

mainly involves the incorporation of endothelial cells or vascular-like structures into a scaffold

and then to implant this scaffold at the desired anatomical site. This approach could favor a

link between the existing vasculature from the host and the endothelialized tissue construct.

58

Page 74: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Furthermore, it could accelerate the formation of functional microvessels in the interior of an

implant [5,6].

Angiogenesis is a key mechanism involved in the development of new blood vessels as well as

during wound healing [7,8]. Several assays have been described to grow networks of

capillaries and to promote angiogenesis in 3D environment, often composed of gel [9-12].

Although some steps of the angiogenesis process have been identified, the origin and

mechanisms involved in lumen formation has been the subject of some debates [13]. The term

lumen is sometimes used to describe features composed of cells forming circular structures in

a nearly 2D plane rather than 3D tube-like structures made of multiple and interconnected

cells, as also suggested by others [14].

Other approaches have been developed to modulate angiogenesis with the use of pre-

vascularized scaffolds made of either natural fibres [15,16] or polymer fibres [17]. Some cells

can orient and often migrate along fibres axis. Cells can also bridge from one fibre to another,

if fibres are separated by distances small enough [18-21]. The fibre-to-fibre distance and the

pattern by which fibres are presented to cells can be adjusted to modulate cell guidance [19-

21]. Furthermore, as shown in a previous study, fibres pre-coated with cell-adhesive

molecules, such as the RGD peptide, were used to guide cell responses [21].

The objective of this study was to develop and validate a culture process that can be

subsequently scaled to bioreactor cultures and that uses polymer fibres to guide the

development of microvessels. We hypothesize that the orientation of microvessels would

subsequently facilitate the flow induction within microvessels lumen for microvascularization

and angiogenesis studies.

3.3 Materials and Methods

3.3.1 Materials

100-um diameter monofilaments made of poly(ethylene terephthalate) (PET, ES305910, Good

Fellow, Devon, USA) were used and fixed onto polycarbonate (Boedeker Plastics Inc., Texas,

USA) frames that fitted into wells of 6-well plates traditionally used in cell culture. The

59

Page 75: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

frames were 32.50 mm in diameter and 3.00 mm thick (Fig. 3.1b). A scanning electron

microscopy (SEM) picture of a PET fibre is presented as Supplementary Figure 3.i.

The phosphate buffered saline solution (PBS IX, pH 7.4) used in these experiments was

prepared from Milli-Q water and NaCl (0.137 M), KC1 (0.003 M), Na2HP04 (0.008 M), and

KH2P04 (0.002 M). This 150 mM solution was diluted in Milli-Q water to make a 10 mM

solution. Hank's balances salt solution (HBSS, H1387) and albumin from bovine serum (BSA,

CAS 9048-46-8) were purchased from Sigma-Aldrich Inc. (St Louis, USA).

3.3.2 Cell culture

Human umbilical vein endothelial cells (HUVECs) were purchased from Cambrex (cc-2519,

Walkersville, MD USA). HUVECs were cultured at 37°C and 5% C0 2 in Ml99 culture

medium (M5017, Sigma-Aldrich) containing 2.2 mg mL"1 sodium bicarbonate (Fisher, Fair

Lawn, USA), 90 \ig mL"1 sodium heparin (HI027, Sigma-Aldrich), 100 U/100 ug mL"1

penicillin/streptomycin (15140-122, Invitrogen Corporation, NY, Grand Island, USA), 10%

fetal bovine serum (FBS, F1051, Sigma-Aldrich), 2 mM L-glutamine (25030149, Invitrogen

Corporation), and 15 (Xg mL"1 endothelial growth factor supplement (ECGS, 356006, BD

Biosciences, San Jose, CA, USA). HUVECs between passages 2 and 6 were used in all

experiments.

3.3.3 PET fibres and fibrin gel preparation

PET fibres were fixed on frames (Fig. 3.1a and 3.1b) to be precisely distanced from each

others (Fig. 3.1c). Frames bearing the fibres were cleaned in RBS (RBS Detergen 35, Pierce

Biotechnology, Rockfdrd, IL, cat 27952), sonicated for 15 min, rinsed with a flow of Milli-Q

gradient water (Millipore Canada, Nepean, Canada) with a resistivity of 18.2 Mflcm, and

finally blow-dried with 0.2-um filter-sterilized air. Frames bearing the fibres were then

immersed in 2 mL of sterile PBS containing 10% antibiotics (100 U/100 ug mL"'

penicillin/streptomycin) and exposed under a UV light for 30 min, as the final sterilization

step.

60

Page 76: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In 6-well plates, fibrin gels were prepared to be used as the attachment bench, using 1 mL/well

of fibrinogen solution (2.0 mg mU1) in HBSS and supplemented with 350 KIU mL1 of

aprotinin. This solution was mixed directly with 1 mL of a thrombin solution (1 U mL"1 in

HBSS) for the polymerization process of fibrinogen into fibrin (5 min at room temperature

followed by 10-20 min at 37°C and 5 % C02).

3.3.4 Cell adhesion assay

After the polymerization process, sterile frames bearing the fibres were transferred to the top

of this fibrin gel. Then, 100,000 cells were seeded directly over the frames bearing the fibres

for cell adhesion. Two (2) mL of Ml99 medium was finally added to each well to cover the

frames (see Fig. 3.Id).

Cell adhesion was investigated after 4 hrs of incubation, as described elsewhere [21-23]. To

count the number of cells attached on fibres surfaces, culture media were removed from the

wells and the frames bearing fibres were incubated 1 hr with Hoechst 33258 (cat. 23491-45-4,

Sigma-Aldrich) at a dilution of 1:10,000 in PBS containing 20% (wt/v) BSA.

Samples were observed under an inverted microscope (Eclipse TE 2000-S, Nikon Corp.,

Chiyoda-ku, Tokyo, Japan) using fluorescence and phase contrast imaging. Images were taken

with a 10X lens in conjunction with a digital CCD camera (Regita 1300R, QIMAGING,

Burnaby, Canada) and an imaging software (SimplePCI, Compix Inc., Minneapolis, MN). The

numbers of nuclei per fibre, corresponding to the number of attached cells, were counted

manually after 4 hrs and 48 hrs following cell adhesion.

3.3.5 Cell culture in 3D environment

After the adhesion phase, frames bearing fibres were transferred to a new gel with the same

composition as that described above, and then covered with a second layer of fibrin that

contained 50,000 HUVECs per milliliter suspended in heparin-free Ml99 medium. The

fibrinogen solution was allowed to clot for 5 min at room temperature and then, at 37°C and

5% CO2 for 10-20 min. After the polymerization process, 2 mL of Ml99 culture medium were

61

Page 77: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

poured over it. Culture media were changed daily and the culture process was evaluated after 2

and 4 days (Figs. 3.1e and 3.If).

3.3.6 Visualization of microvessels

To observe endothelial cell behaviour, samples were observed daily under phase contrast

microscopy and images were recorded.

For cell staining, cell-seeded fibres were gently washed with PBS (3 times) and fixed in a

formaldehyde solution (3.75%, wt/v) in PBS for 20 min. Following three washes with PBS,

cells adhered on fibres were permeabilized with a Triton X-100 solution (0.5% v/v in PBS) for

5 min. Samples were finally rinsed three times in PBS, incubated 1 hr in a PBS solution

containing 20% (wt/v) BSA, and rinsed three times in PBS. Samples were incubated in a

solution containing a mixture of TRITC-phalloidin (1:300 dilution, cat. P1951, Sigma-

Aldrich) and Hoechst 33258 (1:10,000 dilution) made in a blocking buffer solution containing

BSA (20% (wt/v) in PBS) for 1 hr at room temperature, in the dark, and rinsed three times

with PBS and conserved in PBS.

In addition, cells were stained with Dil-Ac-LDL (BT-902, Biomedical Technologies Inc.,

Stoughton, MA, USA) after 2 days of culture to analyze the formation of tube-like structures

and to verify their phenotype.

The number of cell connections was manually quantified after 2 and 4 days of culture by

counting the numbers of sprouts and branch points. Sprout is an elongated structure where a

connection starts, whereas branch points were defined as areas where a single trunk structure

gave rise to two divergent outgrowths.

3.3.7 Images and image processing

Fibres were imaged with an objective of 10X and recorded as high-resolution files (*.tif) to

identify the effects of fibre-to-fibre distance over microvessels formation. An image processing

software (Scion Image; Scion, Frederick, MD) and Picasa2 were used to improve image

quality. These images were used to quantify the number of connections, which originated from

62

Page 78: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

the newly formed tube-like connections along fibres axis. The numbers of connections per

fibre surface area as well as those per fibre length were reported.

3.3.8 Statistical analysis

The results were expressed as mean values + standard deviations for each group of samples (n

= 12), and after assessment of significant differences by one-way variance analysis

(ANOVA), the level of significance was considered at ap-value < 0.05 or < 0.01.

3.3.9 Confocal microscopy imaging of micro vessels

To analyze microvessels between adjacent fibres, frames bearing fibres embedded in fibrin

were gently washed with PBS (3 times) and fixed 20 min in a formaldehyde solution (3.75%,

wt/v, PBS). Following three washes with PBS, cells were permeabilized with a Triton X-100

solution (0.5% v/v in PBS) for 5 min. Samples were finally rinsed three times in PBS,

incubated 1 hr in a PBS solution containing 20% (wt/v) BSA, and rinsed three times in PBS.

Samples were incubated in a solution containing a mixture of TRITC-phalloidin (1:300

dilution) made in a blocking buffer solution containing BSA (20% (wt/v) in PBS) for 1 hr at

room temperature, in the dark, and rinsed three times with Milli-Q water. Finally, samples

were incubated with SYTOX Green Nucleic Acid Stain (1:20,000 dilution in Milli-Q water,

cat. S7020, Invitrogen) for 20 min and washed three times with Milli-Q water.

Images of microvessels were taken with a Confocal Laser Scanning Biological Microscope

(Olympus Fluoview FV300, Olympus Optical Co. Ltd. Tokyo, Japan) equipped with an

Olympus 1X70 camera and recorded as high-resolution and as layer-by-layer files. A complete

image reconstruction was made to illustrate microvessels in a 3D fashion. Images were edited

with the Image-Pro Plus Software to identify microvessels and lumen formation along fibres

axis.

63

Page 79: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

3.4 Results

3.4.1 Cell adhesion on untreated PET fibres

To better promote cell adhesion on untreated PET fibres, frames bearing fibres were placed on

the top of a fibrin gel and HUVECs were then seeded directly over it (Fig. 3.Id). To evaluate

cell adhesion and cell spreading, fibre frames were transferred to a new tissue culture

poly(styrene) (TCPS) plate (Fig. 3.1e) in order to label cell nuclei. To induce the development

of microvessels, HUVEC-covered PET fibres were then sandwiched in fibrin gel; the lower

gel (i.e., that onto which fibre frames were deposited) was prepared without HUVECs, while

the upper fibrin gel (i.e., that covering fibre frames) was seeded with HUVECs (Fig. 3.If).

HUVECs began to attach to untreated PET fibres surfaces after 4 hrs, as indicated by arrows

in Figure 3.2a. The culture was continued for a period of 2 days in order to allow cells to

proliferate, migrate, and spread along the fibres curvature and mostly at the interface between

the fibre and the fibrin gel, as indicated by arrow heads in Figure 3.2b.

Cell density was compared 4 hrs (Fig. 3.2c) and 2 days (Fig. 3.2d) following the initial

incubation. To observe cell orientation and cell spreading along fibres surfaces following 2

days of culture, HUVECs were double-stained with TRITC-phalloidin for actin filaments and

with Hoechst 33258 for nuclei. The epifluorescence microscopy picture (Fig. 3.2e) revealed

that actin filaments were well elongated along fibres axis.

Cell adhesion on fibres is presented as the mean number of attached cells per fibre surface area

(number of cells/mm2) (Fig. 3.2f). The number of anchored cells per fibre surface area

increased as the fibre-to-fibre distance decreased as well as it increased with time. After 4 hrs

of incubation, the number of cells per fibre surface area was significantly (p<0.01) higher with

a fibre-to-fibre distance of 0.05 mm (179 ± 25 cells/mm ) when compared to fibre-to-fibre

distances of 0.1 mm (148 + 14 cells/mm2), 0.15 mm (99 ±11 cells/mm2), and 0.2 mm (68 ± 13

cells/mm2). However, following 2 days of culture, the mean numbers of anchored cells per

fibre surface area were on par with fibres distanced by 0.05 mm (473 ± 48 cells/mm2) and 0.1

mm (455 ± 6 1 cells/mm ), whereas the mean numbers of cells per surface area for fibres

64

Page 80: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

distanced either by 0.15 mm or 0.2 mm were significantly (p<0.01) lowered with values of

257 ± 70 cells/mm2 and 132 + 24 cells/mm2, respectively.

3.4.2 Effect of fibre-to-fibre distance over cell connections and microvessels

formation

When HUVEC-covered fibres separated from each others by 0.05 mm were transferred to new

TCPS plates to be sandwiched in fibrin gel, within 4 hrs some HUVECs formed connection

bridges between adjacent fibres, while for the larger fibre-to-fibre distances tested here, cells

were more elongated along the fibres (Supplementary Figure 3.ii). HUVECs digested fibrin

along fibres. With fibres distanced by 0.1 mm, cell-to-cell connections attached to either one

or two fibres adjacent from each others were formed (Supplementary Figure 3.ii and Fig. 3.3).

When fibres were distanced by 0.15 or 0.2 mm, HUVECs needed more time to develop

connections; even if some connections were found after 2 days, the density was much lower

(Figs. 3.3c and 3.3d).

Phase contrast microscopy pictures of HUVECs labeled with Dil-acetylated-LDL showed a

good cell growth over the fibre curvature and the stability of the cells phenotype over time

(Supplementary Figure 3.ii). Also, testing with Factor VIII at day 2 gives a positive labeling

for endothelial cells (Supplementary Figure 3.ii).

The mean numbers of cell connections between fibres distanced either by 0.05, 0.1, 0.15, or

0.2 mm were compared for periods of 2 and 4 days (Fig. 3.3 and Table 3.1). The mean number

of cell connections found between fibres distanced by 0.1 mm over 2 days (Fig. 3.3c) was

larger than those of fibres distanced either by 0.05, 0.15, or 0.2 mm. A similar trend was

observed after 4 days (Fig. 3.3d).

Cell connections along and in between fibres as well as cell numbers per fibre surface area are

presented in Table 3.1 as mean values + standard deviations. The numbers of connections per

fibre length (number of connections/mm of fibre) after 2 days of culture were on par for fibres

distanced by 0.05 mm (6.4 connections/mm) and 0.1 mm (7.0 connections/mm), whereas it

significantly (p<0.01) decreased with fibres distanced by 0.15 mm (2.6 connections/mm) and

65

Page 81: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

0.2 mm (0.9 connections/mm). Following 4 days, the number of connections per fibre length

for fibres distanced by 0.1 mm (12.1 connections/mm) was significantly (p<0.01) larger than

those of fibres distanced either by 0.05 mm (8.1 connections/mm), 0.15 mm (5.6

connections/mm), or 0.2 mm (2.1 connections/mm) (Table 3.1).

The number of connections per fibre surface area (number of connections/mm2) after 2 and 4

days of culture was significantly (p<0.01) larger for the 0.1 mm fibre-to-fibre distance than

those of fibre-to-fibre distances of 0.05, 0.15 and 0.2 mm (Table 3.1).

The number of cells per fibre surface area increased when fibre-to-fibre distance decreased as

well as when the culture period increased. After 2 days of culture, the number of cells per fibre

surface area for fibres separated by 0.05 mm (338 cells/mm ) was significantly (p<0.01) larger

than those of fibres distanced by 0.1 mm (261 cells/mmz), 0.15 mm (167 cells/mm ), and 0.2

mm (108 cells/mm2). After 4 days, the number of cells per fibre length was also significantly

(p<0.05) larger for a fibre-to-fibre distance of 0.05 mm (460 cells/mm2), when compared to

those of 0.2 mm (133 cells/mm2) and 0.1 mm (332 cells/mm2), which the latter was on par

with that of 0.15 mm (321 cells/mm2) (Table 3.1).

3.4.3 Microvessels formation

The confirmation of fully formed microvessels was verified by confocal microscopy for fibres

distanced by 0.1 mm, from layer-by-layer confocal pictures (Figs. 3.4a-3.4e) and from the

complete image reconstruction. The complete image reconstruction (Fig. 3.4f) reveals the

existence of single lumen in between adjacent fibres. The higher magnification image (Fig.

3.4g) more clearly shows the presence of a lumen inside the network of cell connections

(between upper and lower connections). The picture of microvessels taken using an inverted

microscope is also shown, as comparison (Fig. 3.4h).

In this study, we have successfully imaged microvessels with confocal microscopy without the

need to section the gel containing the cells and the fibres. In fact, histological analyses

following embedding and sectioning were not possible because the fibres delaminated during

sectioning, resulting in microvessels damage (data not shown).

66

Page 82: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

3.5 Discussion

Although cell adhesion on PET surface is limited or even negligible [21,24] since the surface

is moderately wettable (contact angle of approximately 75 degrees) [25], some procedures

have been successfully developed to improve PET fibres biocompatibility [21]. The use of a

fibrin gel to promote the ability of HUVECs to attach, proliferate, migrate, and spread along

PET fibres surface has been successfully demonstrated in the present study. Our findings are

in good agreement with those of Khang et al.[18] that tested fibroblasts adhesion on PET

fibres surfaces. Also, Hadjizadeh et al.[21] and Mirenghi et al.[24] show that it was possible to

enhance human endothelial cell adhesion on plasma-modified and RGD-covered PET

surfaces.

Comparing to a previous study by our group in which PET fibres were surface modified by

plasma polymerization, the number of attached cells per fibre surface area obtained in the

present study was larger. The better cell adhesion on untreated PET fibres observed with the

method described here, as compared to other studies, can be explained in the following ways:

• The fibrin gel can stimulate cell responses because it contains several binding motifs

such as the RGD binding peptides [21-23], and fibrin plasma proteins that interact with

fibronectin to serve as a provisional extracellular matrix (ECM) for cell adhesion

[26,27].

• Fibres offer rigid interfaces to the cells, and as cells generally prefer rigid surfaces than

soft ones, they will migrate to that interface [28-30].

• The surface free energy provided by the bottom fibrin gel favors cell adhesion [31].

Angiogenesis is referred to as the growth of new microvessels from pre-existing blood vessels.

During angiogenesis, endothelial cells orient and organize themselves into tube-like structures

with the presence of lumens, allowing connections of neighboring cells to subsequently form a

network of tubes and initiate new blood vessels [7,14,32]. The angiogenesis process can be

divided into four stages, which conduct to the formation of microvessels [31]: stage 1, cells

digest their surrounding matrix; stage 2, committed cells proliferate, migrate, and

communicate with each others in order to form tube-like connections; stage 3, sprouting and

lumen formation; stage 4, development of a microvessel.

67

Page 83: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

In our system, it can be hypothesized that microvessels formation occurred in the following

steps. Early after HUVEC-covered fibres were embedded in the fibrin gel, spread cells

detached from some areas of the fibres and migrate to the fibrin gel layer, while other

detached cells died. Within 4 hrs after the cell-covered fibres were sandwiched between fibrin

gels, it is hypothesized that cells started to digest fibrin along fibres axis. Afterwards, cells

were able to proliferate and migrate along fibres. Carrying the culture up to 24 hrs, cells on

fibres and those inside fibrin gel interacted and built cell connections. During this phase, cell

cytoskeleton shape became more flatten and elongated to form tube-like structures. The

development of tube-like cell connections was followed by the sprouting and branching of cell

connections until 2 days of culture to form a lumen. The development of tube-like structures

and lumen formation may be associated with a balance between cell migration activity and cell

death, as also reported by other researchers [33,34].

When the culture process was carried out up to 4 days, cells proliferated and migrated even

more, as indicated by the increasing numbers of cell connections and cells per fibre surface

area. Finally, cells formed one tube-like structure composed of multiple cells bridged between

two adjacent fibres, as shown by confocal microscopy pictures (Figs. 3.4f-3.4g). This finding

suggests that the direction of microvessels formation can be guided by the physical

interactions involved between cells and fibres to form a multi-cellular wall, allowing lumen

formation. These microvessels that form between adjacent fibres distanced by 0.1 mm were

associated by morphological events of the differentiation of endothelial cells into tube-like

structures, to subsequently create mature lumen, as also reported by others [14,34,35]. A

complete schematic illustration of the proposed microvascularization development in the

sandwich fibrin gel system containing PET fibres is shown in Figure 3.5 [36].

The fibre-to-fibre distance is an important parameter affecting the stimulation of cell adhesion

as well as microvessels formation. In the adhesion phase, as compared to the larger distances

tested here, smaller fibre-to-fibre distances give more possibility for HUVECs to sail over the

gel to better attach to the rigid fibres surfaces. In this study, endothelial cells were placed at

the interface between the rigid PET fibres and the soft fibrin gel. HUVECs tend to migrate and

68

Page 84: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

attach to the fibres, in particular, integrin cytoplasmic regions of the cells will convert

mechanical forces into biochemical signaling in a process called durotaxis [28,29].

When HUVEC-covered fibres separated by smaller distances (i.e., 0.05 and 0.1 mm) were

sandwiched between two fibrin gels, within 4 hrs cells migrated to form bridges between the

fibres, stabilizing their cytoskeleton, and subsequently building connection strands. In the

larger fibre-to-fibre distances tested in this study (i.e., 0.15 and 0.2 mm), cells required more

time to form and stabilize their connection strands. The numbers of attached cells on the fibres

as well as that of cell connections play an important role in the development of lumen and

micro vessels, as also reported by others [35].

3.6 Conclusions

This study reported the development and validation of a cell adhesion method that uses fibrin

gel to increase HUVECs adhesion and spreading on untreated PET fibres. When fibres pre-

covered with HUVECs were sandwiched between two fibrin gels, microvessels formation

occurred along the fibres. The fibre-to-fibre distance was found to have a significant role in

promoting cell adhesion as well as cell connections between adjacent fibres, thus guiding

microvessels formation along fibres in a 3-dimensional environment. Microvessels formed

large tube-like structures (diameter of approximately 100 urn) between adjacent fibres

distanced by 0.1 mm, after 4 days of culture. When fibres were separated by 0.05 mm, the

distance was too small, so that connections lacked orientation and failed to form a large tube­

like microvessel. In the larger fibre-to-fibre distances (i.e., 0.15 and 0.2 mm), cell connection

strands were rare, so microvessels did not form between adjacent fibres. The requirements of

this culture process were dictated by the possibility to scale the system to bioreactor cultures,

which is the next step in our research program. This proof-of-concept study opens the door to

other experiments in which we plan to study by using this culture process the effects of cell

number, fibre .diameter, ECM density and type, cell types, and the presence of angiogenic

growth factors on microvessels formation. Also, the functionality of these microvessels will be

investigated.

69

Page 85: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

This system can be applied to culture vascularized tissues such as in the development of a

bioartificial pancreas device. These well oriented microvessels can serve to supply nutrients to

growing cells. If the fibres need to be removed from the constructed tissue before use or

implantation, PET fibres can be replaced by biodegradable fibres.

Acknowledgements

This work was supported by the Canadian Foundation for Innovation through an On-going

New Opportunities Fund (project no.7918), the National Sciences and Engineering Research

Council of Canada (NSERC), and by the Universite de Sherbrooke. Mr. Sukmana was

supported by the IDB Merit Scholarship Program. The authors are also grateful to Yannick

Laplante, Mark G. Couture, Gilles Grondin, and Aftab Ahamed for their technical support.

70

Page 86: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

References

1. Sipe JD. Tissue engineering and reparative medicine. Ann NY Acad Sci 2002;961:1-9.

2. Fuchs JR, Nasseri, BA, Vacanti, JP. Tissue engineering: a 21st century solution to surgical

reconstruction. Ann Thorac Surg 2001;72:577-591.

3. Mooney DJ, Sano K, Kaufmann PM, Majahod K, Schloo B, Vacanti JP, Langer R. Long-

term engraftment of hepatocytes transplanted on biodegradable polymer sponges. J

Biomed Mater Res 1997;37:413-420.

4. Atala A. Tissue engineering and regenerative medicine: concepts for clinical application.

Rejuvenation Res 2004;7:15-31.

5. Godbey WT, Atala A. In vitro systems for tissue engineering. Ann NY Acad Sc

2002;961:10-26.

6. Neuman T, Nicholson BS, Sanders JE. Tissue engineering of perfused microvessels.

Microvasc Res 2003;66:59-67.

7. Folkman J, Haudenschild C. Angiogenesis in vitro. Nature 1980;288:551-556.

8. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med 2000;6:389-395.

9. Arthur WT, Vernon RB, Sage EH, Reed MJ. Growth factors reverse the impaired sprouting

of microvessels from aged mice. Microvasc Res 1998;55:260-270.

10. Vernon RB, Sage EH. A novel, quantitative model for study of endothelial cell migration

and sprout formation within three-dimensional collagen matrices. Microvasc Res

1990;57:118-133.

11. Chalupowicz DG, Chowdhury ZA, Bach TL, Martinez J. Fibrin II induces endothelial cell

capillary tube formation. J Cell Biol 1995;130:207-215.

12. Fournier N, Doillon CJ. In vitro angiogenesis in fibrin matrices containing fibronectin or

hyaluronic acid. Cell Biol Int Rep 1992;16:1251-1263.

13. Egginton S, Gerritsen M. Lumen formation in vivo versus in vitro observations.

Microcirculation 2003;10:45-61.

14. Nakatsu MN, Sainson RC, Aoto JN, Taylor KL, Aitkenhead M, Perez-del-Pulgar S,

Carpenter PM, Hughes CC. 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.

71

Page 87: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

15. Unger RE, Peters K, Wolf M, Motta A, Migliaresi C, Kirkpatrick CJ. Endothelialization of

a non-woven silk fibroin net for use in tissue engineering: growth and gene regulation of

human endothelial cells. Biomaterials 2004;25:5137-5146.

16. Fuchs S, Motta A, Migliaresi C, Kirkpatrick CJ. Outgrowth endothelial cells isolated and

expanded from human peripheral blood progenitor cells as a potential source of autologous

cells for endothelialization of silk fibroin biomaterials. Biomaterials 2006;27:5399-5408.

17. Neumann TMD, Hauschka SD, Sanders JE. Tissue engineering of skeletal muscle using

polymer fiber arrays. Tissue Eng 2003;9:995-1003.

18. Khang G, Lee SJ, Lee JH, Lee HB. Interaction of fibroblast cells onto fibers with different

diameter. Korean Polymer Journal 1999;7:102-107.

19. Curtis A, Wilkinson C. Topographical control of cells. Biomaterials 1997;18:1573-1583.

20. Curtis A, Riehle M. Tissue engineering: the biophysical background. Phys Med Biol

2001;46:R47-R65.

21. Hadjizadeh A, Doillon CJ, Vermette P. Bioactive polymer fibers to direct endothelial cell

growth in a three-dimensional environment. Biomacromolecules 2007;8:864-873.

22. Maheshwari G, Brown G, Lauffenburger DA, Wells A, Griffith LG. Cell adhesion and

motility depend on nanoscale RGD clustering. J Cell Sci 2000;113:1677-1686.

23. Hersel U, Dahmen C, Kessler H. RGD modified polymers: biomaterials for stimulated cell

adhesion and beyond. Biomaterials 2003;24:4385-4415.

24. Mirenghi L, Ramires PA, Pentassuglia RE, Rotolo P, Romito A. Growth of human

endothelial cells on plasma-treated polyethyleneterephthalate surfaces. Journal of

Materials Science: Materials in Medicine 2000; 11:327-331.

25. Inoue S, Hori Y, Hirano Y, Inamoto T, Tabata Y. Effect of culture substrate and fibroblast

growth factor addition on the proliferation and differentiation of human adipo-stromal

cells. J Biomater Sci Polymer Edn 2005; 16: 57-77.

26. Makogonenko E, Tsurupa G, Ingham K, Medved L. Interaction of fibrin(ogen) with

fibronectin: further characterization and localization of the fibronectin-binding site.

Biochemistry 2002;41:7907-7913.

27. Tawil B. Fibrin and its applications. In: Guelcher SA, Hollinger JO, editors. An

introduction to biomaterials. Boca Raton FL: CRC Taylor & Francis Press; 2006. p 105-

120.

72

Page 88: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

28. Engler A, Bacakova L, Newman C, Hategan A, Griffin M, Discher D. Substrate

compliance versus ligand density in cell on gel responses. Biophys J 2004;86:617-628.

29. Lo CM, Wang HB, Dembo M, Wang YL. Cell movement is guided by the rigidity of the

substrate. Biophys J 2000;79:144-152.

30. Gray DS, Tien J, Chen CS. Repositioning of cells by mechanotaxis on surfaces with

micropatterned Young's modulus. J Biomed Mater Res A 2003;66:605-614.

31. Schakenraad JM. Cells: their surfaces and interactions with materials. In: Ratner BD,

Hoffman AS, Schoen FJ, Lemond JE, editors. Biomaterials science an introduction to

materials in medicine. San Diego: Academic Press; 1996. p 141-164.

32. Cockerill GW, Gamble JR, Vadas MA. Angiogenesis: model and modulators. Int Rev

Cytol 1995;159:113-160.

33. Bishop ET, Bell GT, Bloor S, Broom U, Hendry NFK, Wheatley DN. An in vitro model of

angiogenesis: basic features. Angiogenesis 1999;3:335-344.

34. Meyer GT, Matthias LJ, Noack L, Vadas MA, Gamble JR. Lumen formation during

angiogenesis in vitro involves phagocytic activity, formation and secretion of vacuoles,

cell death, and capillary tube remodelling by different populations of endothelial cells.

AnatRec 1997;249:327-340.

35. Chrobak KM, Potter DR, Tien J. Formation of perfused, functional microvascular tubes in

vitro. Microvascular Res 2006;71:185-196.

36. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular micro-

environments for morphogenesis in tissue engineering. Nat Biotechnol 2005;23:47-55.

73

Page 89: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Table 3.1: Effects of fibre-to-fibre distance on cells.

Parameters

Number of cell connections per fibre length (connections/mm) Number of cell connections per fibre surface area (connections/mm2) Number of cells per fibre surface area (cells/mm2)

Culture time

(days)

2

4

2

4

2

4

Fibre-to-fibre distances (mm)

0.05

6.4a

±0.6 8.1b

±2.3 23.8" ±2.1 25.8b

±3.1 338a

±26 460a

±157

0.1

7.0a

±1.8 12.1a

±1.5 28.2a

±7.3 42.8a

±11.3 261" ±18 332b

±52

0.15

2.6b

±0.7 5.6C

±1.7 9.1c

±3.7 14.5C

±2.6 167c

±31 321b

±60

0.2

0.9C

±0.3 2.1d

±0.2 2.4d

±1.1 4.1d

±1.3 108d

±32 133c

±58

CD value

p<0.05

1.41

1.91

5.84

6.78

40.08

125.67

p<0.01

1.99

2.69

8.25

9.57

56.56

177.34

*Means in a row followed by different letters are significantly different.

74

Page 90: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

/ P5T

W

Seeding HUVECs

Fibrin gel

Tissue culture poly(styrene) (TCPS) plate

PET fibre HUVEC-covered PET fibre

Fibrin gel + HUVECs

Frame

HUVEC-covered PET fibre

Figure 3.1: Schematic illustration of the in vitro method designed to guide and orient

microvessel formation.

Polycarbonate frames were produced by micro-machining (a and b) to maintain fibres parallel from each others by well defined distances (c). A layer of fibrin gel was prepared to serve as a support for endothelial cells (i.e., HUVECs) adhesion. Polycarbonate frames bearing PET fibres were placed over the gel and HUVECs were seeded directly over fibres and M199 culture medium was added to cover the fibres (d). The culture period was allowed to proceed until HUVECs attached and covered fibres surfaces. When cells reached confluence, frames bearing fibres were transferred to a new plate for further culture (e). PET fibres covered with HUVECs were sandwiched between two fibrin gels and culture medium was added (f); only the upper gel contained HUVECs.

75

Page 91: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

N ^

J

0.05 0.1 0.15 0.2 Fibre-to-fibre distances (mm)

Figure 3.2: HUVECs attachment on PET fibres.

HUVECs attached to fibres after 4 hrs (Step Id in Figure 3.1), as shown by arrows (a). PET fibres were used to guide HUVECs proliferation along fibres axis until 48 hrs, as shown by arrowheads (b). To count the number of attached cells, nuclei were stained with Hoechst after 4 hrs (c) and 48 hrs (d). Actin filaments and nuclei were stained with TRITC-phalloidin and Hoechst, respectively (e). The number of attached cells on fibres surfaces increased as the fibre-to-fibre distance decreased and as the time progressed (f). Scale bars correspond to 100 urn

76

Page 92: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Fibre-to-fibre distances

0.05mm

2 days 4 days

" :* fibre

0.1mm

0.15mm

0.2mm

Figure 3.3: Effect of fibre-to-fibre distance over cell connections and micro vessel

development.

HUVECs built cell connections, as observed by phase contrast microscopy by day 2 (left-hand column) as well as by day 4 (right-hand column). Actin filaments (red) and nuclei (blue) were stained with TRITC-phalloidin and Hoechst, respectively. Fibres were distanced by 0.05 mm (a and b), 0.1 mm (c and d), 0.15 mm (e and f), and 0.2 mm (g and h). Scale bars correspond to 100 |am.

77

Page 93: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

•• Microvessel formation between fibres. •*• (A) Upper connection. •*• (B) Lower connection. •*• (C) Lumen formation between

upper and lower connections.

Figure 3.4: Confocal and inverted microscopy pictures.

After 4 days of culture, cells were stained for actin filaments, using TRITC-phalloidin, and counterstained with SYTOX Green to highlight the position of the nuclei during confocal analyses. Confocal pictures were recorded layer-by-layer from the top to the bottom of samples (a-e). Complete image reconstructions are presented in (f) and (g) for a higher magnification of the lumen. A picture of microvessels taken using an inverted microscope is also shown, as comparison (h). Scale bars correspond to 100 urn.

78

Page 94: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

\5

(

2nd gel S£\ \ +HUVECs ^ \ ~"

/ Bottom gel

HUVECs

HUVECs attachment, migration and elongation on fibres. y view.

HUVEC-eovered PET fibres are sandwiched between fibrin gel. During polymerization process of the fibrin gel, new HUVECs positioned themselves in all levels of the gel down to the first bottom gel.

Fibre \

Fibre 1 ja—r-JJSSsffi" I "*i2

PET fibres

100 um

HUVECs start to make connections with each others between adjacent fibres. Cells become more flatten and elongated and form tube-like structures. y view.

Cell connections and lumen formation between 2 fibres. x view.

Branching, sprouting. y view.

Microvessels formed between adjacent fibres. y view.

Figure 3.5: Schematic illustration of the microvascularization process.

Adapted from Lutolf et al. [36].

79

Page 95: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

SSI' 4 E ''••

3 TJHdB*

Supplementary Figure 3.i: Scanning electron microscopy (SEM) image of a PET fibre.

80

Page 96: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Supplementary Figure 3.ii: Progression of cell connections and microvessel reorganization

along and in between adjacent fibres sandwiched between two fibrin gels.

When fibres were distanced by 0.5mm, HUVECs started to communicate with each others and were able to develop a connection bridge between two adjacent fibres after 4 hrs, as shown by arrows (a). At 0.1 mm fibre-to-fibre distance, cell connections were formed between fibres axis after 24hrs, as shown by discontinued arrow (b). Whereas in larger fibre-to-fibre distances (0.15 and 0.2 mm), connections were rare and discontinued (c and d). Phase contrast microscopy revealed a positive phenotype for endothelial cells (Dil-acetylated LDL uptake), which were distributed along fibres (e). Immuno-labelling with Factor VIII antibody also underlined the positive phenotype of endothelial cells (f). Scale bars correspond to lOOjtm.

81

Page 97: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

CHAPTER 4

The Effects of Co-culture with Fibroblasts and

Angiogenic Growth Factors on Microvascular

Maturation and Multi-cellular Lumen Formation

in HUVEC-oriented Polymer Fibre Constructs

Irza Sukmana1'2, Patrick Vermette1'2

1. Laboratoire de Bioingenierie et de Biophysique de l'Universite de Sherbrooke,

Department of Chemical and Biotechnological Engineering, Universite de Sherbrooke,

2500, blvd de l'Universite, Sherbrooke, QC, Canada, J1K 2R1.

2. Research Centre on Aging, Institut universitaire de geriatrie de Sherbrooke, 1036, rue

Belvedere Sud, Sherbrooke, QC, Canada, J1H 4C4.

Article published in Biomaterials 31 (2010), p. 5100-5109.

DOI: 10.1016/j.biomaterials.2010.02.076

http://www.sciencedirect.com/science/iournal/01429612

82

Page 98: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

4.1 Abstract

In the present study, polymer monofilaments were embedded in fibrin seeded with human

umbilical vein endothelial cells (HUVEC) to guide HUVEC attachment and migration in order

to form oriented vessel-like structures between adjacent monofilaments. Histology and

fluorescent fibrin experiments confirmed that microvessel-like structures, which were

developing between polymer monofilaments embedded in fibrin, contained a lumen. The

effect of human fibroblasts and growth factors (VEGF and bFGF) over the microvessel

formation process was tested. The effects of VEGF and bFGF were dose dependent. The effect

of VEGF was optimum at the lower concentration tested (2ng/mL), while that of bFGF was

optimum at the higher tested concentration (20ng/mL). Furthermore, the use of fibroblasts

significantly improved the maturation of the microvessels compared to control and to samples

cultured with VEGF and bFGF.

4.2 Introduction

Microvascularization is a process in which endothelial cells form microvessels. It is involved

in the progression of cancerous cell masses [1-3]. Also, functional microvessels are necessary

to supply nutrients and remove wastes in cell and/or tissue aggregates in order to improve

tissue substitute survival and function [3,4], therefore justifying the development and

application of methods to support microvessel formation.

Angiogenesis is a multistage process involving the sprouting of blood vessels from pre­

existing vasculature [2,4]. Briefly, endothelial cells degrade the extracellular matrix followed

by cell proliferation, migration, and cell-cell interactions leading to the formation of tube-like

structures, and eventually to sprouting and lumen formation with adjacent vessels [4,5].

Several in vitro and in vivo angiogenesis assays have been developed to study and follow the

development of sprouts and lumens and to validate the efficacy of both pro- and anti-

angiogenic therapeutics [5,6]. In vitro assays include endothelial cells grown in different gels

[6,7], microcarrier bead assays [8,9], and aortic rings embedded in gels [10-12], only to name

a few. In vivo assays include the chick chorioallantoic membrane (CAM) [13], the Matrigel

implant [14], and the retinal angiogenesis assay [15].

83

Page 99: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

A number of reports have shown that pericytes [16] and smooth muscle cells [3] can maintain

the patent tubular structure of blood vessels, and at the same time provide important factors to

improve the maturation of microvessels [16,17]. Fibroblasts have been found to improve

capillary sprout growth and stabilization in collagen and fibrin gels [16-18]. At some stage of

the angiogenesis process, smooth muscle cells inhibit endothelial cell proliferation and new

vessel formation, implying that mural cells interact with endothelial cells to stabilize newly

formed capillaries [19].

On a different note, other studies reported that angiogenic growth factors, such as vascular

endothelial growth factor (VEGF) [17,20] and basic fibroblast growth factor (bFGF) are

essential for the induction of angiogenesis [21,22]. However, there are some contradictory

data on how VEGF and bFGF concentrations influence cellular responses [12,17].

Although some aspects of angiogenesis, including endothelial cell migration, proliferation and

phenotype differentiation seem to be established among the scientific community, many

studies fail to model the maturation of microvessels up to the stage of lumen formation and

even just to prove the existence of such multi-cellular lumen [17]. In addition, often

microvessel networks are randomly distributed in a dispersing gel environment, and lack any

patterning and specific orientation [22,23]. In tissue engineering applications and cancer

research, the modeling and study of directional sprouting and microvessel formation is

important [23,24]. Firstly, we believe that by orienting the development of microvessels,

induction of fluid flow within the lumen of these microvessels will be facilitated [24].

Secondly, having a system in which microvessels can be patterned opens the door to new in

vitro models of metastasis progression in cancer research [23-26] to better understand how

biosignalling localization affects microvessel development in a 3D environment.

For these reasons, here we have further optimized and validated our in vitro model of

microvessel patterning using polymer monofilaments embedded in fibrin. In a previous study,

we have reported the development of an angiogenesis assay in which microvascularization

process was modulated in a directional fashion [24]. It was demonstrated that polymer fibres

deposited on top of a fibrin gel allowed cell attachment on these monofilaments. Furthermore,

84

Page 100: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

when HUVEC-covered fibres were subsequently sandwiched in fibrin, cells established

connections with each others, thus leading to the formation of one microvessel between two

adjacent fibres. Using this culture process, the aims of the present study were to investigate the

effect of human fibroblasts and two angiogenic growth factors (i.e., vascular endothelial

growth factor (VEGF) and basic fibroblast growth factor (bFGF)) on microvessel maturation

and to assess lumen formation.

4.3 Materials and Methods

4.3.1 Materials

100-um diameter poly(ethylene terephthalate) monofilaments (PET, ES 305910, Good Fellow,

Devon, USA) were fixed onto polycarbonate (Boedeker Plastics Inc., Texas, USA) frames that

fitted into traditional 6-well plates used in cell culture. The frames were 22.5 mm in diameter

and 2.0 mm thick [24].

The phosphate buffered saline solution (PBS IX, pH 7.4) used in these experiments was

prepared from Milli-Q water and NaCl (0.137 M), KC1 (0.003 M), Na2HP04 (0.008 M), and

KH2P04 (0.002 M). This 150 mM solution was diluted in Milli-Q water to make a 10 mM

solution. Hank's balances salt solution (HBSS, H1387) and albumin from bovine serum (BSA,

CAS 9048-46-8) were purchased from Sigma-Aldrich Inc. (ON, Canada).

Vascular endothelial growth factor (VEGF) used here is a human recombinant VEGFi65

with a 38.2-kDa disulfide-linked homodimeric protein consisting of two 165 amino acid

polypeptide chains. Basic fibroblast growth factor (bFGF) is a human recombinant with a

17.2-kDa protein consisting of 154 amino acid residues. VEGF165 (cat.# 100-20) as well as

bFGF (cat.# 100-18B) were purchased from PeproTech (PeproTech inc., NJ, USA).

4.3.2 Cell culture

Human umbilical vein endothelial cells (HUVEC) were purchased from PromoCell (C-12200,

Heidelberg, Germany). HUVEC were cultured at 37°C and 5% C02 in Ml99 culture medium

(M5017, Sigma-Aldrich) supplemented with 2.2 mg mL"1 sodium bicarbonate (Fisher, Fair

85

Page 101: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Lawn, USA), 90 ug ml/1 sodium heparin (H1027, Sigma-Aldrich), 100 U/100 ug mL"1

penicillin/streptomycin (15140-122, Invitrogen Corporation, Grand Island, NY, USA), 10%

foetal bovine serum (FBS, F1051, Sigma-Aldrich), 2 mM L-glutamine (25030149, Invitrogen

Corporation), and 15 ug mL"1 endothelial growth factor supplement (ECGS, 356006, BD

Biosciences, San Jose, CA, USA). HUVEC between passages 2 and 6 were used in all

experiments. Skin fibroblasts extracted from human foreskin and purchased from PromoCell

(C-12350, Heidelberg, Germany) were cultured in Ml99 supplemented with 10% FBS and

100 U/100 ug mL"1 penicillin/streptomycin at 37°C and 5% CO2. Fibroblasts between

passages 6 and 15 were used in all experiments.

4.3.3 Culture system and fibrin preparation

PET fibres were fixed on polycarbonate frames to be precisely distanced from each other [24].

Frames bearing fibres were cleaned in RBS detergent 35 (cat 27952, Pierce Biotechnology,

Rockford, IL), sonicated for 15 min, rinsed with a flow of Milli-Q gradient water (Millipore

Canada, Nepean, Canada) with a resistivity of 18.2 Mii-cm, and finally blow-dried with 0.2-

um filter-sterilized air. Frames bearing fibres were then immersed in 2 mL of sterile PBS

containing 10% antibiotics (100 U/100 ug mL"1 penicillin/streptomycin) and exposed under

UV light for 30 min, as the final sterilization step.

In 6-well plates, fibrin gels were prepared to be used as the attachment bench using 1 mL/well

of fibrinogen solution (4.0 mg mL"1) made in HBSS and supplemented with 350 KIU mL"1 of

aprotinin. This solution was directly mixed with 1 mL of a thrombin solution (2 U mL"1 in

HBSS) for the polymerization process of fibrinogen into fibrin, 5 min at room temperature

followed by 20 min at 37°C and 5 % C02 .

4.3.4 Cell adhesion and angiogenesis assay

After the polymerization process, sterile frames bearing the fibres were transferred and

deposited on the top of a fibrin gel prepared as described in Section 2.3. Then, 100,000

HUVEC were directly seeded over it and cultures were run for 2 days to allow cell attachment.

Two (2) mL of supplemented Ml99 medium were finally added to each well to cover the

frames [24].

86

Page 102: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

After the attachment phase, frames bearing fibres were transferred to a new fibrin gel with the

same composition as described above, and then covered with a second layer of fibrin that

contained 100,000 HUVEC per milliliter suspended in heparin-free Ml99 medium. The

fibrinogen solution was allowed to clot for 5 min at room temperature and then, at 37°C and

5% CO2 for 30 min. After the polymerization process, 2 mL of M199 culture medium (with or

without growth factors) were poured over it. Culture media were changed daily and the culture

process was evaluated after 2 and 4 days. For the co-culture experiment, 300,000 of human

foreskin fibroblasts were mixed directly with Ml99 culture media and added on the top of the

fibrin. In another set of experiments, HUVEC were fed with M199 supplemented with either

VEGF or bFGF (2, 5 or 20 ng/mL) with no fibroblasts.

4.3.5 Visualization of microvessels and lumen formation

To observe endothelial cell behaviour, pictures were daily taken with a phase contrast

microscope. After 2 and 4 days, the fibrin gels containing the frames were fixed for further

examination. For cell staining, cell-seeded fibres were gently washed with PBS (3 times) and

fixed in a formaldehyde solution (3.75%, wt/v) in PBS for 20 min. Following three washes

with PBS, cells adhered on fibres were permeabilized with a Triton X-100 solution (0.5% v/v

in PBS) for 15 min. Samples were finally rinsed three times in PBS, incubated 1 h in a PBS

solution containing 2% (wt/v) BSA, and rinsed three more times in PBS. Samples were

incubated in a solution containing a mixture of rhodamine-phalloidin (1:300 dilution, R415,

Molecular Probes, Eugene, OR) for F-actin labelling and Hoechst 33258 (1:10,000 dilution,

cat. # B2883, Sigma-Aldrich) for nuclei staining, made in a blocking buffer solution

containing BSA (2% (wt/v) in PBS) for 1 h at room temperature, in the dark. Finally, after

three washes with PBS, samples were conserved with 3 mL per well of PBS.

Samples were imaged and recorded as high-resolution files (*.tif). To improve quality, images

were then processed with Image-Pro-Plus Software. The number of cell-cell connections was

manually quantified after 2 and 4 days of culture by counting the numbers of sprouts and

branch points. Sprout is an elongated structure where a connection starts, whereas branch

87

Page 103: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

points are single trunk structures that give rise to two divergent outgrowths. The numbers of

cells as well as cell-cell connections per fibre length are reported.

To assess the formation of multi-cellular lumen, human fibrinogen conjugated with Alexa

Fluor 546 (F-13192, Molecular Probes, Eugene, OR) was combined with unlabelled human

fibrinogen at a mass ratio of 1:10 (conjugated to unconjugated fibrinogen) to produce the fibrin

gel, as in Section 2.3, in which the HUVEC-covered fibres were sandwiched. In this

experiment, HUVEC were labelled with the CellTracer™ CFSE staining (C34554, Molecular

Probes, Eugene, OR) for 30 min prior to the experiment. The fibrin degradation by HUVEC

could then be examined using confocal microscopy.

In another series of experiments, fibres covered with unlabelled HUVEC were sandwiched

between fluorescent fibrin (see the above paragraph for composition) and after polymerization,

fibroblasts were subsequently added on the top of the fibrin gel. The cultures were carried out

for a period of 4 days. Following fixation and permeabilization, as described above, cells were

stained with Alexa Fluor 488 green phalloidin (1:300 dilution, A12379, Molecular Probes,

Eugene, OR) for F-actin and Hoechst for nuclei. Layer-by-layer images were taken with a

Confocal Laser Scanning Biological Microscope (Olympus Fluoview FV1000, Olympus

Optical Co. Ltd., Tokyo, Japan).

4.3.6 Histological section

Fibrin gels containing fibre holders were fixed overnight in 4% neutral buffered formalin.

Fibres embedded in fibrin were gently removed from their frame, and processed for paraffin

sections according to standard protocols [27]. Six-um-thick sections were then prepared for

hematoxylin and eosin staining (both from Sigma-Aldrich). Pictures were taken with the

bright field mode of the Biolmaging Navigator fluorescence microscope (Olympus FSX100,

Olympus Optical Co. Ltd., Tokyo, Japan).

4.3.7 Imaging microvessels by confocal microscopy

To investigate microvessel formation between adjacent fibres, frames bearing fibres embedded

in fibrin were gently washed with PBS (3 times) and fixed 20 min in a formaldehyde solution

88

Page 104: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

(3.75%, wt/v, PBS). Following three washes with PBS, cells were permeabilized with a Triton

X-100 solution (0.5% v/v in PBS) for 5 min. Samples were rinsed three times in PBS,

incubated 1 h in a PBS solution containing 2% (wt/v) BSA, and rinsed three more times in

PBS. Samples were then incubated in a solution of rhodamine-phalloidin red (1:300 dilution)

made from a blocking buffer containing BSA (2% (wt/v) in PBS) for 1 h at room temperature,

in the dark, and rinsed three times with PBS. Finally, samples were incubated with SYTOX

Green Nucleic Acid Stain (cat. S7020, 1:10,000 dilution in Milli-Q water, Invitrogen) for 20

min and washed three times with PBS.

Images of microvessels were taken with a Confocal Laser Scanning Biological Microscope

(Olympus Fluoview FV1000, Olympus Optical Co. Ltd., Tokyo, Japan) equipped with an

Olympus 1X70 camera and recorded as high-resolution layer-by-layer files. Images were edited

with Olympus FV10-ASW 2.0 viewer and a complete image reconstruction was made to

provide a 3D view of the microvessels.

4.3.8 Statistical analysis

The results are expressed as mean values ± standard deviations for each group of samples.

Data were statistically analyzed by using the analysis of variance (ANOVA).

4.4 Results and Discussion

This three-dimensional cell culture system allows to precisely controlling the fibre-to-fibre

distance to 100|im [24]. When HUVEC-covered fibres were embedded in fibrin also

containing HUVEC, cells oriented and elongated along the fibre axis at the fibre-gel interface,

allowing the patterning of microvessels. When using HUVEC-free monofilaments embedded

in fibrin containing HUVEC, neither tube-like structures nor cell-cell connections were

observed (Supplementary Figure 4.i). The step-by-step process of microvessel development

occurring in the culture system described here has been presented elsewhere [24].

Compared to our previous study in which lower HUVEC concentration was used in the fibrin

gel [24], the numbers of cells as well as cell-cell connections per fibre length after 2 and 4

days of culture found in this present study were higher. In the present study, the cell density

89

Page 105: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

was increased, as we found that it plays a significance role in the microvessel development.

This is in agreement with another study revealing that when cell number was smaller than a

certain threshold value, the formation of tube-like cell connections, sprouting and branching

were not observed [28].

After sandwiching the HUVEC-covered fibres in fibrin and adding fibroblasts, some cells that

cover the fibres migrate along the fibre axis and some into the fibrin (Fig. 4.1a) in order to

interact with the cells located inside the gel, finally forming cell-cell connections after 1 day of

culture. During this phase, cell cytoskeleton shape became more flatten and elongated to form

tube-like structures (discontinued arrows, Fig. 4.1b) and even some cells were able to bridge

between two fibres to build a cell connection (thick white arrows, Fig. 4.1b). When HUVEC

were cultured in the absence of fibroblasts, many HUVEC migrated away from the fibres and

failed to build cell-cell connections, and even though connections were formed after 2 days

(Fig. 4.1c), the number of connections was lower when compared to the system in which

fibroblasts were added (Fig. 4.Id).

Lower magnification image of samples with fibroblasts reveals the tube-like structures seen

between adjacent fibres, almost completely covering the area between fibres after 4 days (Fig.

4.1e). Also, a complex tube-like structure network occurred between adjacent fibres (black

and white arrows, Figs. 4.If and 4.1g).

When HUVEC-covered fibres were co-cultured with fibroblasts for a period of 4 days,

confocal microscopy pictures (Figs. 4.2a-4.2e) confirmed the better maturation of the

microvessels, compared to control samples (Supplementary Figure 4.ii). A lower magnification

of the reconstructed confocal image (Fig. 4.2f) as well as a higher magnification image (Fig.

4.2g) also revealed that the fibrin between adjacent fibres, where cells formed connections,

was degraded over time. In these experiments (Figs. 4.2f and 4.2g), fibrin was prepared using

fibrinogen conjugated with a red fluorescent probe mixed with regular fibrinogen. HUVEC

were labelled with CFSE CellTracer™ (green).

90

Page 106: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

The effect of VEGF and bFGF over the formation of cell-cell connections and micro vessel

development was dose-dependent (Table 4.1). Numbers of cell-cell connections between

fibres as well as cell numbers per fibre length are listed in Table 4.1. The mean numbers of

cell-cell connections between fibres as well as cell numbers per fibre length between controls

and systems with either VEGF or bFGF at three concentrations (i.e. 2, 5 and 20ng/mL) were

compared for a culture period of 2 days (Figs. 4.3a-4.3d and Table 4.1). The effect of VEGF

was stronger at 2 and 5ng/mL (Fig. 4.3a), when compared to the highest concentration tested

here (20ng/mL, Fig. 4.3b) and to the control system with no added growth factors. The

addition of bFGF yielded higher numbers of cell-cell connections and cells per fibre length at

a concentration of 20ng/mL (Fig. 4.3d), when compared to those with bFGF concentrations of

2 or 5ng/mL (Fig. 4.3c) and control.

Our results (Fig. 4.3 and Table 4.1) showed that VEGF and bFGF supported cell proliferation

and migration, thus enhancing the formation of cell-cell connections in a dose-dependent

manner. This finding is in good agreement with other studies [17,22,29-31]. The use of VEGF

at a concentration of 20ng/mL resulted in lower cell numbers and cell-cell connections

compared to control cultures. This phenomenon is referred to as the biphasic effect of VEGF,

as also reported by others [30,32].

Following 2 and 4 days of culture, the number of cell-cell connections per fibre length

(connections/mm of fibre) was statistically significantly higher when fibroblasts were added

compared to those of control and samples with bFGF (20ng/mL) and VEGF (2ng/mL). Similar

findings were found when the numbers of cells per fibre length (cells/mm of fibre) were

compared for the different samples (Table 4.2).

Cell-cell connections and evidence of the degradation of fibrin are presented in Figure 4.4.

Examining the layer-by-layer confocal pictures, multiple cell-cell connections were observed

(see left sides of Figs. 4.4a-4.4d). At time zero, cells were rounded with a strong uniform

signal from the fluorescent fibrin (Fig. 4.4f). Cell-cell connections (discontinued lines) were

associated to areas where fibrin was degraded (stars, see right sides of Figs. 4.4a-4.4d). We

believe that these cell-cell connections are made of multiple cells resulting in lumen, as

91

Page 107: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

reported by others [33,34]. The complete image reconstruction (Fig. 4.4e) also reveals that the

multi-cellular connections covered almost all the area in between the adjacent fibres.

Histological sections also confirmed that HUVEC were forming multi-cellular lumen (star,

Fig. 4.4g). Lumen were guided by the PET fibre (discontinued arrow, Fig. 4.4h) and made up

of multiple cells (arrows, Fig. 4.4i).

These findings reveal that fibroblasts enhance microvessel formation, development and

maturation. This is in agreement with several studies available in the scientific literature. For

example, fibroblasts were found to stabilize endothelial cell-lined tube formation [34], and

provide important signalling that lead to the maturation of capillaries made of HUVEC

[32,35]. Fibroblasts are known to release many growth factors including VEGF, FGF, platelet-

derived growth factor (PDGF), and angiopoietin-1 [12,16-18,35]. When comparing the effects

of VEGF and bFGF and fibroblasts over the formation of cell-cell connections and subsequent

microvessel maturation, our results show that fibroblasts can provide sufficient pro-angiogenic

factors to promote vessel-like formation (Figs. 4.2a-4.2e). The use of fibroblasts yielded

higher numbers of cells and cell-cell connections, when compared to experiments involving

VEGF or bFGF. However, there are still some debate on how VEGF and bFGF concentration

affects angiogenesis and microvascularization (see Table 4.3 for some examples of results

from the scientific community).

In samples with fibroblasts, they were deposited on top of the upper fibrin gel. In this way,

they were from a distance of approximately 2 to 3 mm from the endothelial cells and the

fibres. To ascertain that fibroblasts were not migrating into the fibrin and so would never come

in direct contact with HUVEC up to 4 days of culture, in some experiments fibroblasts were

labelled with the CFSE CellTracer to follow their localization (Supplementary Figure 4.iii).

These experiments reveal that fibroblasts remain on the top of the fibrin and thus the effect of

fibroblasts over microvessel development was not a result of a direct contact between

fibroblasts and HUVEC.

To carry out longer cultures (more than 4 days), we believe that the recruitment of fibroblasts

or pericytes at the site of microvessel formation will be necessary in order to stabilise and

92

Page 108: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

completely close the walls of the microvessels formed between adjacent fibres, as also

postulated by some researchers [34,36-38]. For instance, many studies have demonstrated the

ability of pericytes, an immature cell type that can differentiate into fibroblasts, smooth

muscle cells or macrophages, to initiate vascular sprouts, to stabilize vessels and to support

vessel maturation [18,39,40].

4.5 Conclusions

The development of a functional micro-vasculature is a crucial factor for the survival as well

as the development and function of engineered vascularised tissue substitutes. Here, the effects

of fibroblasts and two growth factors (VEGF and bFGF) over microvessel development were

studied using a system in which human umbilical vein endothelial cells (HUVEC) are guided

by polymer monofilaments to form oriented vessel-like structures. VEGF and bFGF were

shown to enhance HUVEC proliferation and migration, thus improving the development of

cell-cell connections and subsequent microvessel development. When HUVEC-covered fibres

were co-cultured with fibroblasts, the HUVEC responses were more pronounced compared to

control and to cultures in which VEGF and bFGF were used. The use of fibroblasts resulted in

more mature microvessels containing more cell-cell connections. These microvessel-like

structures were also shown to contain a lumen. This culture process in which microvessels

with multi-cellular lumen are patterned can find applications in cancer and tissue engineering

studies.

Acknowledgements

This work was supported by the Canadian Foundation for Innovation/Ministere de 1'Education

du Quebec through an On-going New Opportunities Fund (project no.7918), the National

Sciences and Engineering Research Council of Canada (NSERC), and by the Universite de

Sherbrooke. Mr. Sukmana was supported by the IDB Merit Scholarship Program. The authors

are grateful to Johanne Dussault and Dr. Sameh Geha, for their help in histology, and to Gilles

Grondin and Dr. Leonid Volkov for their assistance in confocal microscopy analysis. Also, the

authors would like to thank Evan Dubiel and Georges Sabra for their critical review of the

manuscript.

93

Page 109: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

References

1. Szoke T, Kayser K, Baumhakel JD, Trojan I, Furak J, Tiszlavicz L, et al. Prognostic

significance of microvascularization in case of operated lung cancer. Eur J Cardiothorac

Surg 2005;27:1106-1111.

2. Folkman J, Haudenschild C. Angiogenesis in vitro. Nature 1980;288:551-556.

3. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med 2000;6:389-395.

4. Neumann T, Nicholson BS, Sanders JE. Tissue engineering of perfused microvessels.

Microvasc Res 2003;66:59-67.

5. Cockerill GW, Gamble JR, Vadas MA. Angiogenesis: model and modulators. Int Rev

Cytol 1995;159:113-160.

6. Passaniti A, Taylor RM, Pili R, Guo Y, Long PV, Haney JA, et al. A simple, quantitative

method for assessing angiogenesis and antiangiogenic agents using reconstituted basement

membrane, heparin, and fibroblast growth factor. Lab Invest 1992;67:519-528.

7. Bayless KJ, Davis DE. Sphingosine-1-phosphate markedly induces matrix

metalloproteinase and integrin-dependent human endothelial cell invasion and lumen

formation in three-dimensional collagen and fibrin matrices. Biochem Biophys Res

Commun 2003;312:903-913.

8. Nakatsu MN, Hughes CC. Optimized three-dimensional in vitro model for the analysis of

angiogenesis. Methods Enzymol 2008;443:65-82.

9. 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.

10. Nicosia RF, Ottinetti A. Growth of microvessels in serum-free matrix culture of rat aorta:

A quantitative assay of angiogenesis in vitro. Lab Invest 1990;63:115-122.

ll.Blacher S, Devy L, Burbridge MF, Roland G, Tucker G, Noel A, et al. Improved

quantification of angiogenesis in the rat aortic ring assay. Angiogenesis 2001;4:133-142.

12. Zhu WH, Iurlaro M, Maclntyre A, Fogel E, Nicosia RF. The mouse aorta model: influence

of genetic background and aging on bFGF- and VEGF-induced angiogenic sprouting.

Angiogenesis 2003;6:193-199.

13.Forough R, Wang X, Martinez-Lemus LA, Thomas D, Sun Z, Motamed K, et al. Cell-

based and direct gene transfer-induce angiogenesis via a secreted chimeric fibroblast

94

Page 110: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

growth factor-1 (sp-FGF-1) in the chick chorioallantoic membrane (CAM). Angiogenesis

2003;6:47-54.

14. Akhtar N, Dickerson EB, Auerbach R. The sponge/matrigel angiogenesis assay.

Angiogenesis 2002;5:75-80.

15.Uemura A, Kusuhara S, Katsuta H, Nishikawa S. Angiogenesis in the mouse retina: a

model system for experimental manipulation. Exp Cell Res 2006;312:676-683.

16. Nehls V, Denzer K, Drenckhahn D. Pericyte involvement in capillary sprouting during

angiogenesis in situ. Cell Tissue Res 1992;270:469-474.

17. Nakatsu MN, Sainson RCA, 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.

18. Velazquez OC, Snyder R, Liu ZF, Fairman RM, Herlyn M. Fibroblast-dependent

differentiation of human microvascular endothelial cells into capillary-like 3-dimensional

networks. FASEB J 2002;16:1316-1318.

19. Darland DC, D'Amore PA. TGF/? is required for the formation of capillary-like structures

in three-dimensional cocultures of 10T1/2 and endothelial cells. Angiogenesis 2001 ;4:11-

20.

20. Taylor KL, Henderson AM, Hughes CC. Notch activation during endothelial cell network

formation in vitro targets the basic HLH transcription factor HESR-1 down regulates

VEGFR-2/KDR expression. Microvasc Res 2002;64:372-383.

21. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific

growth factors and blood vessel formation. Nature 2000;407:242-248.

22. Nehls V, Herrmann R, Huhnken M. Guided migration as a novel mechanism of capillary

network remodeling is regulated by basic fibroblast growth factor. Histochem Cell Biol

1998;109:319-329.

23. Eichmann A, Noble FL, Autiero M, Carmeliet P. Guidance of vascular and neural network

formation. Current Opinion in Neurobiology 2005; 15:108-115.

24. Sukmana I, Vermette P. Polymer fibres as contact guidance to orient microvascularization

in a 3D environment. J Biomed Mater Res A 2010;92:1587-1597.

25. Zetter BR. Angiogenesis and tumor metastasis. Annu Rev Med 1998;49:407-424.

95

Page 111: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

26. Folkman J. Seminars in Medicine of the Beth Israel Hospital, Boston. Clinical application

of research on angiogenesis. N Engl J Med 1995;333:1757-1763.

27. Fisher AH, Jacobson KA, Rose J, Zeller R. Preparation of cell tissues for fluorescence

microscopy. In: Spector DL, Goldman RD, editors. Basic methods in microscopy.

Woodbury: Cold Spring Harbor Lab Press, 2006.p. 105-123.

28. Martineau L, Doillon CJ. Angiogenic response of endothelial cells seeded dispersed versus

on beads in fibrin gels. Angiogenesis 2007;10:269-277.

29. Hutchings H, Ortega N, Plouet J. Extracellular matrix-bound vascular endothelial growth

factor promotes endothelial cell adhesion, migration, and survival through integrin

ligation. FASEB J 2003;17:1520-1522.

30. Nakatsu MN, Sainson RCA, Perez-del-Pulgar S, Aoto JN, Aitkenhead M, Taylor KL, et al.

VEGFm and VEGF165 regulate blood vessel diameter through vascular endothelial growth

factor receptor 2 in an in vitro angiogenesis model. Lab Invest 2003;83:1873-1885.

31. Nillesen STM, Geutjes PJ, Wismans R, Schalkwijk J, Daamen WF, van Kuppevelt TH.

Increased angiogenesis and blood vessel maturation in acellular collagen-heparin scaffolds

containing both FGF2 and VEGF. Biomaterials 2007;28:1123-1131.

32. Chen Z, Htay A, Santos WD, Gillies GT, Fillmore HL, Sholley MM, et al. In vitro

angiogenesis by human umbilical vein endothelial cells (HUVEC) induced by three-

dimensional co-culture with glioblastoma cells. J Neurooncol 2009;92:121-128.

33. Allt G, Lawrenson JG. Pericytes: cell biology and pathology. Cells Tissues Organs

2001;169:1-11.

34. Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circ Res

2005;97:512-523.

35. Chen X, Aledia AS, Ghajar CM, Griffith CK, Putman AJ, Hughes CCW, et al.

Prevascularization of a fibrin-based tissue construct accelerates the formation of functional

anastomosis with host vasculatures. Tissue Eng Part A 2009;15:1363-1371.

36. Hellstrom M, Gerhardt H, Kalen M, Li X, Eriksson U, Wolburg H, et al. Lack of pericytes

leads to endothelial hyperplasia and abnormal vascular morphogenesis. J Cell Biol

2001;153:543-553.

96

Page 112: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

37. Davis GE, Sanger WB. Endothelial extracellular matrix: biosynthesis, remodeling, and

functions during vascular morphogenesis and neovessel stabilization. Circ Res

2005;97:1093-1107.

38. Moon JJ, West JL. Vascularization of engineered tissues: approaches to promote

angiogenesis in biomaterials. Curr Top Med Chem 2008;8:300-310.

39. Jain RK. Molecular regulation of vessel maturation. Nat Med 2003;9:685-693.

40. Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat

Rev Mol Cell Biol 2007;8:464-478.

41. Uriel S, Brey EM, Greisler HP. Sustained low level of fibroblast growth factor-1 promotes

persistent microvascular network formation. Am J Surg 2006;192:604-609.

42. Xue L, Greisler HP. Angiogenic effect of fibroblast growth factor-1 and vascular

endothelial growth factor and their synergism in a novel in vitro quantitative fibrin-based

3-dimensional angiogenesis system. Surgery 2002;32:259-267.

97

Page 113: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Table 4.1: Effects of VEGF and bFGF concentrations over the numbers of cells and cell-cell

connections following 2 days of culture.

Cell number and cell-cell connection*

Number of cell-cell connections per fibre length (connections/mm) Number of cells per fibre length (cells/mm)

Growth factor concentrations (ng/mL)

Control

6.6d

±0.9

52.6d

±7.7

VEGF

2

9.1a

±0.4

83.9a

±2.3

5

7.2" ±1.1

82.2a

±6.3

20

5.1e

±1.0

53.0 d

±12. 3

bFGF

2

7.0C

±1.1

68.8C

±4.4

5

8.1b

±0.9

78.6b

±5.8

20

9.8a

±1.0

85.8a

±5.4

CD value

p<0.05

0.95

7.07

p<0.01

1.43

10.60

*Means in a row followed by different letters are significantly different.

Table 4.2: Comparison of the effects of fibroblasts, VEGF and bFGF over the numbers of cells

and cell-cell connections.

Cell number and cell-cell connection*

Number of cell-cell connections per fibre length (connections/mm) Number of cells per fibre length (cells/mm)

Culture time

(days)

2

4

2

4

Parameters

Control

6.6C

±0.9 12.5C

±1.6 52.6b

±7.7 92.7C

±18.5

Fibroblasts

12.4a

±0.9 20.9a

±3.8 89.5a

±4.6 124.9a

±12.1

VEGF (2ng/mL)

9.2b

±0.4 13.2C

±2.2 83.9b

±2.3 89.5b

±5.7

bFGF (20ng/mL)

9.8b

±1.0 16.4b

±1.5 85.8b

±5.4 97.9" ±5.3

CD value

p<0.05

0.87

2.64

5.78

12.62

p<0.01

1.35

4.11

8.99

19.65

*Means in a row followed by different letters are significantly different.

98

Page 114: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Table 4.3: Review of some studies on the effects of VEGF and bFGF on endothelial cells and

angiogenesis.

Cell type/culture method

HUVEC cultured in fibrin and collagen gel containing growth factors and sphingosine-1-phosphate(SlP). EC were seeded on microcarrier beads and then embedded in fibrin containing growth factors.

Mouse aortic rings (contained EC and mural cells) were cultured in a collagen-based matrix.

HUVEC were cultured on microcarrier beads and embedded in fibrin, then covered with Ml99 containing growth factors.

EC cultured on microcarrier beads embedded in a serum-free fibrin with bFGF. Human umbilical artery endothelial cells were cultured in plastic wells pre-coated with growth factors. HUVEC were cultured on microcarrier beads, then embedded in fibrin gels and covered with Ml99 containing growth factors. Canine jugular vein EC (CJEC) cultured in fibrin glue then embedded in fibrin gel and covered with Ml99 containing FGF-1. Bovine aorta endothelial cells cultured in fibrin glue, then embedded in fibrin and covered with Ml99 containing 1 or 2 growth factors.

Growth factors

bFGF and VEGF (40ng/mL)

VEGF (lOOng/mL) and bFGF (30ng/mL) VEGF and bFGF (5, 20, and 50ng/mL)

VEGF and bFGF at several concentrations (l-35ng/mL) bFGF (30ng/mL)

VEGF165 and VEGF189 at 4ug/mL

VEGF12i and VEGF165 at several concentrations (l-35ng/mL) FGF-1 1, 10, 100 ng/mL

FGF-1 and VEGF at concentrations between 1 to lOOng/mL.

Results

S IP combined with either VEGF or FGF stimulated EC to invade gels and form tube-like structures and not in the absence of SIP. The use of either bFGF or VEGF stimulated capillary formation from EC.

Both growth factors dose-dependently stimulated angiogenesis, with bFGF showing a stronger effect than VEGF. The stronger stimulatory dose for both factors was 20 ng/mL. VEGF enhanced sprout formation from beads but not bFGF. VEGF was optimal at low concentrations (2.5-5ng/mL) and decreased at higher concentrations (15-35ng/mL). bFGF played an important role in the regulation of guided EC migration during angiogenesis. Immobilized VEGF supported cell migration and survival. VEGF 189 enhanced EC adhesion when compared to VEGF165. Low concentrations of both VEGF12i and VEGFi65 promoted the growth of long, thin vessels, whereas higher concentrations of VEGF remarkably enhanced vessel diameter. Low concentration of FGF-1 (1 ng/mL) maintained a persistent microvascular network, whereas higher concentrations resulted in an abnormal CJEC phenotype and vessel regression. Significant effect of FGF-1 started at 1 ng/mL while VEGF at 2ng/mL. The optimum effect was at higher concentration (lOOng/mL) for both growth factors. When combining bFGF and VEGF at concentration of lOng/mL, the effect was greater than either bFGF or VEGF alone.

Ref.

[7]

[9]

[12]

[17]

[22]

[29]

[30]

[41]

[42]

EC: endothelial cells.

99

Page 115: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Figure 4.1: Effect of fibroblasts over microvessel development.

After being sandwiched in fibrin, HUVEC migrated along the fibre axis and some toward the gel (a). After 1 day of culture, some cells were able to build cell-cell connections along the fibre axis (discontinued arrows) and even able to bridge between fibres (arrows) (b). In the absence of fibroblasts, many cells failed to establish cell-cell connections after 2 days of culture (arrows) (c). Comparing to the control, the use of fibroblasts produced more tube-like cell-cell connections after 2 days (d). After 4 days of culture, low magnification images of samples with fibroblasts reveal that cell-cell connections covered almost all the area between adjacent fibres (e). With the presence of fibroblasts, phase contrast microscopy (f) as well as immunofluorescence (F-actin and nucleus staining) (g) confirmed the presence of a complex tube-like structure network after 4 days of culture. Scale bars correspond to 100 um.

100

Page 116: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Figure 4.2: Immunofluorescence picture illustrating the effect of fibroblasts on

microvessel development.

After 4 days of culture, cells were stained for F-actin using rhodamine-phalloidin (red) and counterstained with SYTOX Green to highlight the position of nuclei during confocal analyses. Confocal pictures were recorded as layer-by-layer images from the top to the bottom of samples (a-d). Complete image reconstruction is presented in (e). To confirm the degradation of fibrin after 4 days, HUVEC were stained with the CFSE Cell Tracer (green) prior to be cultured in fibrin made of a mixture of fluorescent (red) and non-conjugated fibrinogen. A complete image reconstruction is shown at low (f) and high magnification (g). Scale bars correspond to 100 urn.

101

Page 117: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Concentration (ng/mL)

Low (2-5) High (20)

VEGF

bFGF

Figure 4.3: Immunofluorescence picture (F-actin and nuclei staining) illustrating the effect of

VEGF and bFGF on microvessel development. Scale bars correspond to 100 um.

102

Page 118: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

\ .

fibre

AS-

^

t "-.

Figure 4.4: Illustration of multi-cellular lumen formation for cultures carried out for a period

of 4 days with fibroblasts.

HUVEC were stained for F-actin (green) and nuclei (blue). Fibrin was made using a mixture of fibrinogen conjugated with Alexa Fluor 546 (red) and non-conjugated fibrinogen. Microvessel structure and fibrin degradation are illustrated in the layer-by-layer pictures (a-d) as well as in the complete image reconstruction (e). Image of the culture system at time zero (f). Examination of histological sections stained using hematoxylin and eosin reveals HUVEC lining (arrows) and lumen formation (star) (g). Lumens were guided by the fibre (discontinued arrow) (h). Higher magnification reveals that HUVEC are lining (i). Scale bars correspond to 30 um (a-f), to 100 um (g and h), and to 10 um (i).

103

Page 119: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Culture time

8h

48 h

72 h

Uncovered PET fibres

PET fibres pre-covered with HUVEC

^ « ^ | » ! W « i « « * « * » ^ ' # i * | S ' Jffi*s4

wwnipiiiim mn'tfiii'.'imr •aa>***i*,«<t,*m&<c^&i

man, fmtiwi* <am mmxiiMen^mf.i^*^ jfflfc «as*. trsM • R W vwwmijmmmw «•***«*

t a^ f^p^te^ l

I d

••-• - « • -^- rt

f

• i i f f • » « • - • a t ••»•

^ * - ^ t *4 . « , • » • • •

.. * : i . <*uf--<*->* v . •*»

. - * • I , • • • . •• •

• . * » •

.» " *

-^g».*IWi.-Aff4*W :«i.JW'» * f • < * '•#*- * * l

• * * • . ; ! " 7 . . . . - . -V <f * •- ' I - - <

PtTt-hie

! ' *v i5 i ' "it,-- .•ta-.fc.iit.i.'.yi -; ' * . *.*••******-ar-tim. :'*s .9**.ri»i.-"ram

Supplementary Figure 4.i: Comparison between systems using cell-free fibres (left) and

HUVEC-covered fibres (right) over cell-to-cell connections and microvessel development.

Scale bars correspond to 100 um.

104

Page 120: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

P6T fibre

Supplementary Figure 4.ii: Immunofluorescence confocal pictures of a sample cultured with

no fibroblasts.

After 4 days, cells were stained for F-actin using rhodamine-phalloidin and counterstained with SYTOX Green to highlight the position of nuclei. Confocal pictures were recorded as layer-by-layer images from the top to the bottom of samples (a-d). Complete image reconstruction is presented in (e). Scale bars correspond to 100 u.m.

105

Page 121: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Supplementary Figure 4.iii: Fibroblasts stained with the CFSE CellTracer.

106

Page 122: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

CHAPTER 5

Conclusions and Suggestions

5.1 Conclusions

This thesis project aimed at developing a tridimensional cell culture system for the guidance of

microvessel formation. It was the initial part of a larger research program aiming to create an

oriented functional blood microvessel network within three-dimensional scaffolds. The

development of mature and oriented blood microvessels should facilitate oxygen and nutrient

transport as well as waste removal within tissue constructs and can be applied to culture

vascularised tissue substitutes.

Overall, the results presented in this thesis reveal that this novel culture system can be used to

successfully orient the development of microvessels. However, to reach the goals targeted in

this research program, some further investigations should be carried out in the future. For

example, investigating microvessel functionality should be done in future studies, as suggested

by others [Chrobak et al. 2006]. Testing the culture method presented in this thesis in a

dynamic culture system [Chrouinard et al. 2009] to achieve more mature microvessels should

be further studied.

Briefly, this thesis presents the development and validation of a novel cell adhesion method to

increase HUVEC adhesion and spreading on untreated polymer fibres. Further tridimensional

culture experiments using these HUVEC pre-coated fibres sandwiched between fibrin gels

revealed that this system can induce formation of tube-like structures in a directional fashion.

The optimum fibre-to-fibre distance to promote oriented microvessels was found to be IOOUMTI.

The use of growth factor (i.e., Vascular Endothelial Growth Factor (VEGF) and Basic

Fibroblast Growth Factor (bFGF)) was applied to improve microvessel development. Also, co-

culturing these HUVEC-covered fibres with fibroblasts significantly increased the maturation

107

Page 123: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

of the formed microvessels. Further histology and fluorescent fibrin experiments also

confirmed that microvessel-like structures, which were developing between polymer

monofilaments embedded in fibrin, contained a lumen.

We believe that the well oriented microvessels reported in this thesis, which consisted of

multi-cellular lumens, should facilitate flow induction within these lumens. Also, this assay

will find applications in cancer research and tissue engineering.

5.2 Conclusion (en francais)

Ce projet de these visait a developper un systeme tridimensionnel de culture cellulaire pour

orienter la formation de micro-vaisseaux. II represente la partie initiale d'un programme de

recherche plus large qui a pour objectif de creer un reseau de micro-vaisseaux sanguins

fonctionnels a l'interieur d'echafaudages tridimensionnels. Le developpement de micro-

vaisseaux sanguins matures devrait faciliter le transport de l'oxygene et des nutriments ainsi

que l'enlevement des dechets dans les masses tissulaires.

Dans l'ensemble, les resultats presentes dans cette these montrent que ce systeme de culture

novateur peut etre utilise avec succes pour orienter le developpement des micro-vaisseaux.

Cependant, pour atteindre les objectifs vises dans ce programme de recherche, des

investigations complementaires devraient etre menees dans le futur. Par exemple, 1'etude de la

fonctionnalite des micro-vaisseaux devrait etre realisee tel que suggere par d'autres [Chrobak

et al. 2006]. Tester la methode de culture presentee dans cette these dans un systeme de culture

dynamique [Chrouinard et al. 2009] pour atteindre des micro-vaisseaux plus matures devrait

etre fait.

En bref, cette these presente le developpement et la validation d'une nouvelle methode pour

augmenter 1'adherence et la migration de cellules endotheliales humaines provenant de la

veine ombilicale (HUVEC) sur des fibres de polymere non-modifiees. D'autres experiences de

culture tridimensionnelle en utilisant ces fibres pre-recouvertes d'HUVEC et « sandwichees »

entre des gels de fibrine ont revele que ce systeme peut induire la formation de structures

108

Page 124: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

tubulaires de facon directionnelle. La distance optimale fibre-a-fibre pour promouvoir la

formation de micro-vaisseaux orientes est d'environ lOOum.

L'utilisation du facteur de croissance (i.e., Vascular Endothelial Growth Factor (VEGF) et le

basic Fibroblast Growth Factor (bFGF)) a ete appliquee pour ameliorer le developpement des

micro-vaisseaux. De plus, dans une autre serie d'experiences, la co-culture des fibres

recouvertes de HUVEC avec des fibroblastes augmente significativement la maturation des

micro-vaisseaux formes. L'histologie et des experiences menees avec de la fibrine

fluorescente ont egalement confirme que ces pseudo-micro-vaisseaux qui se developpent entre

les fibres de polymere incorporees dans la fibrine possedaient un lumen.

Nous croyons que les micro-vaisseaux bien orientes rapportes dans cette these, qui possedent

des lumens multicellulaires, devraient faciliter l'induction d'un flux au sein de ces lumens. En

outre, cette methode de culture et son utilisation pourrait trouver des applications dans la

recherche sur le cancer et en ingenierie tissulaire.

5.3 Suggestions

The proof-of-concept of this tridimensional culture system opens the door to other

experiments either in static or dynamic cultures using a perfusion bioreactor system in order to

achieve the final goal of this research program. The following future works and techniques are

suggested:

5.3.1 Further studies of the angiogenesis assessment

Angiogenesis is a multi-factorial process in which new blood vessels are formed from existing

vessels [Carmeliet 2000; Folkman and Haudenschild 1980]. The process of angiogenesis

occurring during tumour progression involves, in many respects, events found during

morphogenesis and development [Salcedo et al. 2005]. In tissue engineering, models of

angiogenesis development were adapted for the vascularization of tissue constructs. In this

thesis, the emphasis was to demonstrate the proof-of-concept that microvessel formation can

109

Page 125: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

be guided by polymer monofilaments. Studies to further assess angiogenesis development

using our novel system can include:

• To investigate the involvement of integrins during microvessel formation. Some

integrins, particularly av[33, av[35 and cc5(3l, have been identified as markers for ligand-

based and functional mediators of tumour angiogenesis [Hwang and Varner, 2004; Davis

et al. 2002]. For example, Max et al. (1997) have reported the use of

immunohistochemistry to show that normal tissue gives a lower expression of ocvP3

integrin when compared to tissues undergoing angiogenesis. Also, RGD-containing

peptides, which bind to the ocvP3 integrin, have been proposed as tracer for tumour

targeting and angiogenesis imaging [Haubner et al. 2001].

• To study the expression of growth factors and proteolytic enzymes during

angiogenesis process. The assessment of angiogenesis can also be done by investigating

growth factor expression from endothelial cells (EC), such as Vascular Endothelial

Growth Factor (VEGF), Basic Fibroblast Growth Factor (bFGF), and Platelet-derived

Growth Factor (PDGF) [Nakatsu et al. 2003b; Yancopoulos et al. 2000]. For example,

using Western blot, the level of VEGF expression was compared between control cells and

those undergoing angiogenesis. Also, the expression of proteolytic enzymes (i.e., matrix

metalloproteinases (MMP)) that can be related to fibrin degradation during capillary

formation can indicate when endothelial cells undergo angiogenesis [Koh et al. 2009].

• Proteins as markers for angiogenesis. The occurrence of angiogenesis process can also

assessed by characterizing the expression of other proteins related to endothelial cell

function, such as endothelial nitric oxide synthase (eNOS), vascular cell adhesion

molecule-1 (VCAM-1), von Willebrand Factor (vWF), inter-cellular adhesion molecule-1

(ICAM-1), platelet endothelial cell adhesion molecule-1 (PECAM-1), and E and P-

selectins [Bramfeld et al. 2010; Davis et al. 2002; Montano et al. 2009], only to name

a few.

• Gene expression during the development of an oriented microvessel. Transfection

techniques (i.e., adenoviral and siRNA transfection) have been used to silence (either

knock-out or knock-down) some genes (i.e., Tie-2, delta 4, notch 1 and HESR-1) that are

related to angiogenesis [Nakatsu et al. 2003a; Taylor et al. 2002]. In this study, HUVEC

110

Page 126: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

were firstly treated in order to knock-down the gene which is related to angiogenesis,

before using these cells. Therefore, the proof of angiogenesis process occurring in our 3D

method presented in this thesis can be further investigated.

• To study expression of the extra-domain B (EDB domain) of fibronectin. The EDB

domain is a sequence of 91 amino acids that can be inserted into the fibronectin molecule

[Halin et al. 2001] and is one of the best markers for angiogenesis [Alessi et al, 2004]. In

tumour progression, fibronectin containing EDB accumulates around neovascular

structure, which appears to be found only in tissues undergoing angiogenesis [Halin et al.

2001]. To evaluate the expression, radio-labelled single-chain Fv (scFv) antibody which

would recognise the same epitope of EDB domain of fibronectin can be used. For

example, Viti et al. (1999) have concluded that scFv expression of tumour vasculature was

higher compared to that in normal tissues [Viti et al. 1999].

5.3.2 The effect of dynamic culture system over microvessel development and

proof of microvessel functionality

Dynamic cell culture experiments can be carried out using a simple set-up (see Figure 5.1) as

well as using an automated bioreactor system, which has been previously described

[Chrouinard et al. 2009]. Using a dynamic culture system, below are further suggested works

to study the development of oriented microvessels.

• Effect of dynamic culture over microvessel development. Endothelial cells are

experiencing fluid shear stress and dynamic flow conditions in the entire circulatory system

[Aird 2007]. The use of dynamic culture parameters (i.e., flow rate and pulsation) can allow

different mechanical stimulation [Levenberg et al. 2005]. A simple dynamic culture (see

Figure 5.1) can be used to study the effect of flow rate, while to investigate the influence of

pulsation an automated bioreactor system such as the one available in Prof. Vermette's

laboratory can be used. In this study, the effect of mechanical stimulation on cellular

guidance and microvessel development could be further investigated in order to find the

optimum parameter.

• Assessment of angiogenesis process during microvessel development in a dynamic

culture environment. For further validation of the occurrence of angiogenesis, several

111

Page 127: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

characterizations listed in the above section could be used. For example, the level of VEGF

and PDGF-BB expression on EC cultured with flow were found to be higher than those in

static cultures [Sumpio et al. 1998].

• Study of endothelial nitric oxide (NO) production and eNOS expression in dynamic

culture. As previously described, expression of some proteins can used to follow the state

of the EC involved in the angiogenesis process. Among them eNOS expression and NO

production have been found to increase when endothelial cells were exposed to flow

conditions [Li et al. 2003].

• Smooth muscle oc-actin (SMocA) to assess microvessel maturation. During the

formation of new microvessels in vivo, the expression of SMocA is indicative of the

presence of smooth muscle cells (SMC), which are essential to blood microvessel

maturation [Jain 2003]. Also, dynamic culture with flow and pulsation has been found to

improve the structural organization of SMC [Akhyari et al. 2002; Holtorf et al. 2005].

Therefore, when using co-culture systems of EC with SMC, the level of SMocA expression

at the site of microvessel formation is of significance in the overall angiogenesis process

[Kofidis et al. 2002].

• Assessment of microvessel functionality. After the microvascular tube is being mature

i.e., the vessel wall is completely covered by endothelial cells and pericytes, further

assessment of its functionality is important. Testing its functionality can be adapted from

techniques published by others [Chrobak et al. 2006].

112

Page 128: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Figure 5.1 Simple set-up for dynamic cell cultures.

A custom-made cell culture chamber (a) was prepared to fit the fibre-frame (b). The simple dynamic culture system consists of a power supply (c) (located outside a C02 incubator) and a microperistaltic pump connected to a bottle containing culture medium; air filter, silicon tubing and culture chamber can be placed within the incubator (d). An image of HUVEC cultured with a flow rate of 80 mL/min after 3 days is presented at low (e) and high (f) magnifications, illustrating healthy cells.

113

Page 129: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

References

Aird, W. C. (2007) Phenotypic heterogeneity of the endothelium: II. Representative vascular

beds. Circ.Res. 100, p. 158-173.

Akhyari, P., et al. (2002) Mechanical stretch regimen enhances the formation of bioengineered

autologous cardiac muscle grafts. Circulation 106, p. 1137-1142.

Alessi, P., Ebbinghaus, C. and Neri, D. (2004) Molecular targeting of angiogenesis.

Biochimica et Biophysica Acta 1654, p. 39-49.

Bramfeld, H., et al. (2010) Scaffold vascularisation: a challenge for three-dimensional tissue

engineering. Curr.Med.Chem. Submitted.

Carmeliet, P. (2000) Mechanisms of angiogenesis and arteriogenesis. Nat.Med. 6, p. 389-395.

Chouinard, J. A., et al. (2009) Design and validation of a pulsatile perfusion bioreactor for 3D

high cell density cultures. Biotechnol.Bioeng. 104, p. 1215-1223.

Chrobak, K. M., Potter, D. R. and Tien, J. (2006) Formation of perfused, functional

microvascular tubes in vitro. Microvascular Res. 71, p. 185-196.

Davis, G. E., Bayless, K. J. and Mavila, A. (2002) Molecular basis of endothelial cell

morphogenesis in three-dimensional extracellular matrices. Anat.Rec. 268, p. 252-275.

Folkman, J. and Haudenschild C. (1980) Angiogenesis in vitro. Nature 288, p.551-556.

Halin, C , Zardi, L. And Neri, D. (2001) Antibody-based targeting of angiogenesis. News

Physiol.Sci. 16, p. 191-194.

Haubner, R., et al. (2001) Noninvasive imaging of alpha(v)beta3 integrin expression using

18F-labeled RGD-containing glycopeptides and positron emission tomography. Cancer

Res.6\,p. 1781-1785.

Holtorf, H. L., Jansen, J. A. and Mikos, A. G. (2005) Flow perfusion culture induces the

osteoblastic differentiation of marrow stromal cell-scaffold constructs in the absence of

dexamethasone. J.Biomed.Mater.ResA 72, p. 326-334.

Hwang, R. and Varner, J. (2004) The role of integrins in tumor angiogenesis.

Hematol.Oncol.Clin.N.Am. 18, p. 991-1006.

Kofidis, T., et al. (2002) In vitro engineering of heart muscle: artificial myocardial tissue.

J.Thorac.Cardiovasc.Surg. 124, p. 63-69.

Koh, C. J., et al. (2009) Parthenogenesis-derived multipotent stem cells adapted for tissue

engineering applications. Methods 47, p. 90-97.

114

Page 130: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

Levenberg, S., et al. (2005) Engineering vascularized skeletal muscle tissue. Nat.Biotechnol.

23, p. 879-884.

Li, Y., et al. (2003) Effects of pulsatile sherar stress on nitric oxide production and endothelial

cell nitric oxide synthase expression by ovine fetoplacental artery endothelial cells. Biol.

Reproduction 69, p. 1053-1059.

Max, R., et al. (1997) Immunohistochemical analysis of integrin alpha vbeta3 expression on

tumor-associated vessels of human carnicomas. Int. J.Cancer 71, p. 320-324.

Montano, I., et al. (2009) Formation of human capillaries in vitro: the engineering of

prevascularized matrices. Tissue Eng. Part A 15, p. 1-15.

Nakatsu, M. N., et al. (2003a) 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. 66, p. 102-112.

Nakatsu, M. N., et al. (2003b) VEGF(121) and VEGF(165) regulate blood vessel diameter

through vascular endothelial growth factor receptor 2 in an in vitro angiogenesis model.

Lab.Invest. 83, p. 1873-1885.

Salcedo, X., et al. (2005) Review article: angiogenesis soluble factors as liver disease markers.

Aliment.Pharmacol.Ther. 22, p. 23-30.

Sumpio, B. E., et al. (1998) Regulation of PDGF-B in endothelial cells exposed to cyclic

strain. Arterioscler.Thromb.Vasc.Biol. 18, p. 349-355.

Taylor, K. L., Henderson, A. M. and Hughes, C. C. (2002) Notch activation during endothelial

cell network formation in vitro targets the basic HLH transcription factor HESR-1 and

downregulates VEGFR-2/KDR expression. Microvasc.Res. 64, p. 372-383.

Viti, F., et al. (1999) Increased binding affinity and valence of recombinant antibody

fragments lead to improved targeting of tumoral angiogenesis. Cancer Res. 59, p. 347-

352.

Yancopoulos, G. D., et al. (2000) Vascular-specific growth factors and blood vessel formation.

Nature 407, p. 242-248.

115

Page 131: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

APPENDIX A

Protocols for cell staining

Basic Solutions

• Phosphate buffered saline solution (PBS IX, pH 7.4).

• Formaldehyde solution (3.75%, wt/v) in PBS for fixation.

• Triton X-100 solution (0.5% v/v in PBS) for permeabilization.

• PBS containing 20% (wt/v) BSA (referred to as PBS/BSA below) as a blocking buffer

solution.

A . l Protocol for F-Actin staining

One of the common markers for F-actin filament is TRITC-phalloidin (cat. #P1951, Sigma-

Aldrich). Phalloidin is isolated from Aminata phalloides mushroom and binds in the same site

of F-actin (see Appendix B for detail information).

Step-by-step staining procedure for F-Actin:

• Samples were gently washed with PBS (3 times) then fixed in a formaldehyde solution

for 20 min.

• Following three washes with PBS, samples were then permeabilized with a Triton X-

100 solution for 5 min at room temperature.

• After three rinsing steps in PBS, samples were incubated 1 hour in a PBS/BSA

solution for a blocking process then rinsed three times in PBS.

• Samples were then incubated in a solution containing a mixture of TRITC-phalloidin

(1/300) and Hoechst (1/10,000) made in PBS/BSA for 1 hour at room temperature, in

the dark.

• After two final rinsing steps with PBS, samples were conserved in PBS.

116

Page 132: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

A.2 Protocol for Di l -Ac-LDL uptake

Dil-Ac-LDL (Acetylated Low Density Lipoprotein) is a marker to distinguish endothelial cells

from other cell types (i.e., fibroblasts, smooth muscle cells, epithelial cells and pericytes).

When cells were labelled with Dil-Ac-LDL, the lipoprotein is degraded by lysosomal enzymes

and the fluorescent probe (Dil) accumulates in the intracellular membranes of endothelial

cells. Dil-Ac-LDL used in this project was purchased from Biomedical Technologies Inc.,

Stoughton, MA, USA (cat. # BT-902).

Step-by-step staining procedures:

• Dilute Dil-Ac-LDL to lOug/mL in supplemented Ml99 media.

• Add this diluted staining solution over the endothelial cells monolayers then incubate

at 37°C and 5% C0 2 for 4 hours.

• Remove the Dil-Ac-LDL staining solution and wash twice with Ml99 medium with no

Dil-Ac-LDL.

• After incubation at 37°C and 5% CO2 for 30 minutes, cells are ready to be observed

with fluorescence microscopy using a standard filter for Alexa 546 or rhodamine.

117

Page 133: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

A.3 Protocol for von Willebrand Factor staining

Von Willebrand Factor (vWF) is a large multimeric glycoprotein secreted by megakaryocytes

and endothelial cells. vWF participates fundamentally in the hemostasis process in the

association with thrombogenic Factor VIII (FVIII) (i.e., blood clotting factor or pro-cofactor).

During the hemostasis, vWF and FVIII have been found to be synthesized and released by the

endothelium into the bloodstream. Therefore, vWF is known as a marker for endothelial cells.

Goat anti-vWF was purchased from Haematologic Technologies Inc., USA (cat.# PAHVWF-

G) and diluted 1/500 in PBS/BSA. Secondary antibody FITC rabbit anti-goat was purchased

from Chemicon (cat.# AP106F) diluted 1/100 in PBS/BSA mixed with Hoechst (cat. # H-

33258 Sigma-Aldrich) (1/10000).

Below is the detailed protocol:

• Samples were gently washed with PBS (3 times) then fixed in a formaldehyde solution

for 20 min.

• Following three washes with PBS, samples were permeabilized with a Triton X-100

solution for 5 min at room temperature.

• After three rinsing steps in PBS, samples were saturated in rabbit serum (Biodesign,

USA; cat# N66010R) diluted 1/20 in PBS for 30 minutes, then rinsed three times in

PBS.

• Incubate cells with primary antibody goat anti-vWF in PBS/BSA for 1 hour, then rinse

three times in PBS.

• Incubate samples with secondary antibody FITC rabbit anti-goat and Hoechst in

PBS/BSA for 1 hour at room temperature and protect from light.

• Finally, rinse three times in PBS.

118

Page 134: PJM UNIVERSITY DE 10 SHERBROOKE DEVELOPPEMENT D'UN … · PJM UNIVERSITY DE 10 SHERBROOKE Faculte de genie Departement de genie chimique et de genie biotechnologique DEVELOPPEMENT

APPENDIX B

Cell cytoskeleton

The cytoskeleton is a framework structure inside the cytoplasm of eukaryotic cells. Its

function is mainly to give a cell its distinctive shape and high level of internal organization.

The major structural elements of cytoskeleton are: microtubules, intermediate filaments, and

microfilaments. These three major elements play important roles to support cell movement

and motility.

Microtubules (MT) are the largest structures and are composed of a hollow structure with

outer and inner diameters of 25nm and 15nm, respectively. MT are responsible for motility of

the cell and also support cell shape and distribution of micro and intermediate filaments.

Intermediate filaments (IF) have a diameter about 8-12nm and are the most stable and least

soluble constituents compared to the two other major cytoskeleton components. IF are thought

to have a tension-bearing role since they are often observed in areas of mechanical stress. In

contrast with other components, they differ in size and chemical compositions between one

tissue to another.

Microfilaments (MF) are the smallest structures, with a diameter of approximately 7nm. They

play important role in cell contraction and cell locomotion. They also contribute in the

maintenance of cell shape. MF are composed of the protein actin, called G-actin, which can

form double-helical strands of F-actin filaments (F stands for filamentous). To date, F-actin

filaments are known as sites for cell-cell and cell-ECM interactions.

119