fabrication of a 3-dimensional cardiac tissue using a ... · fabrication of a 3-dimensional cardiac...

134
Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach by Brendan Martin Pue-Bun Leung A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Brendan Martin Pue-Bun Leung 2011

Upload: others

Post on 28-May-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach

by

Brendan Martin Pue-Bun Leung

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Institute of Biomaterials and Biomedical Engineering

University of Toronto

© Copyright by Brendan Martin Pue-Bun Leung 2011

Page 2: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

ii

Fabrication of a 3-dimensional cardiac tissue using a modular

tissue engineering approach

Brendan Martin Pue-Bun Leung Doctor of Philosophy

Institute of Biomaterials and Biomedical Engineering University of Toronto, 2011

Abstract

Implantation of engineered cardiac tissue may restore lost cardiac function to damaged

myocardium. We propose that functional cardiac tissue can be fabricated using a modular,

vascularized tissue engineering approach developed in our laboratory. In this study, rat aortic

endothelial cells (RAEC) were coated onto sub-millimetre size modules embedded with

cardiomyocyte-enriched neonatal rat heart cells (CM) and assembled into a contractile,

macroporous sheet-like construct.

Cell morphologies, contractility and responsiveness to electrical stimulus were examined

to evaluate the function of the resulting modular construct. CM embedded modules contracted

spontaneously at day 7 post-fabrication and remained viable in vitro at day 14. Modules cultured

in 10% bovine serum were more contractile and responsive to external stimulus compared to

10% FBS medium cultured modules. VE-cadherin staining showed a confluent layer of RAEC

on CM embedded co-culture modules at day 7. Co-culture modules were also contractilie, but

when compared to CM only modules their electrical responsiveness was slightly diminished.

Modules assembled into macroporous sheets retained their characteristics at day 10 post-

assembly. Micrographs from histological sections revealed the existence of muscle bundles near

the perimeter of modules and at inter-module junctions.

Page 3: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

iii

The fate of modular cardiac tissues in vivo was examined using two implantation

strategies based on a syngeneic animal model. Co-culture modules (CM and EC) were either

injected into the peri-infarct zone of the heart, or fabricated into a patch form and implanted

over a right ventricular free wall defect. In both models, donor EC were involved in the

formation of blood vessels-like structures, which appeared to have connected with the host

vasculature. Co-culture implants had a higher overall vessel density compared to CM-only

implants, but only in the absence of MatrigelTM. Moreover, donor CM organized into striated

muscle-like structures, at least when MatrigelTM was removed from the matrix. Together these

results suggest that modular cardiac tissue can survive and develop into native-like structures

when implanted in vivo and the potential of the modular approach as a platform for building 3-D

vascularised cardiac tissue should be explored in greater depth.

Page 4: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

iv

Acknowledgements

I’d like to begin by expressing my most sincere gratitude to my supervisor, Dr. Michael

Sefton, for his guidance, patience and understanding during my graduate training. I feel very

blessed to have had Dr. Sefton as my mentor. His genuine interest in my personal and

professional development has shaped me into the researcher that I am today. I will cherish all

the lessons that I have learned from our numerous discussions in his office and our countless

chit-chats in front of the espresso machine; I thank you from the bottom of my heart.

I am also very fortunate to have had a wonderful group of professors who served on my

advisory committee. Thank you Dr. Simmons, Dr. Backx and Dr. Rocheleau for all your

insightful comments and suggestions. In particular, I’d like to thank Dr. Radisic for helping me

get the project off the ground and for all her expert advice in the field of cardiac tissue

engineering.

I’d like to thank everyone in the Sefton lab and the 4th floor of CCBR for all the laughter

and support. A special thanks goes out to Chuen Lo for all his technical assistance and

encouraging words. To Omar Khan, Mark Butler and Rohini Gupta, thank you for making my

graduate experience memorable both inside and outside of the lab.

I am forever indebted to my parents, Robert and Nancy Leung. Thank you for giving me

the inspiration and opportunity to pursue my dreams. Also, to my brother Brian, thank you for

your support and good times.

Finally, to my loving wife Helen, thank you for believing in me. Your love and

encouragement have helped me see the positive side of everything and have made me a better

person. To you, I dedicate this thesis.

Page 5: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

v

Table of Contents

ABSTRACT .................................................................................................................... II

ACKNOWLEDGEMENTS ............................................................................................. IV

TABLE OF CONTENTS ................................................................................................. V

LIST OF FIGURES ...................................................................................................... VIII

LIST OF TABLES ........................................................................................................... X

ABBREVIATIONS ......................................................................................................... XI

CHAPTER 1: RESEARCH INTRODUCTION, HYPOTHESES AND OBJECTIVES ..... 1

1.1 The Need for Engineered Cardiac Tissue ................................................................................................... 1

1.2 Research Overview and Hypothesis ............................................................................................................ 2

1.3 Research Objectives ........................................................................................ Error! Bookmark not defined. 1.3.1 Overall Objectives ...................................................................................................................................... 3 1.3.2 Specific Objective 1: Fabrication of CM-EC co-culture modules ............................................................. 3 1.3.3 Specific Objective 2: Characterization of CM/EC co-culture modules ..................................................... 3 1.3.4 Specific Objective 3: Fate of CM/EC co-culture modules in vivo ............................................................. 4

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

CHAPTER 2: CURRENT METHODS AND CHALLENGES IN CARDIAC TISSUE ENGINEERING ............................................................................................................... 7

2.1 Engineered Cardiac Muscles for Functional Cardiac Regeneration ................................................................ 7

2.2 General Considerations for in vitro Tissue Engineering ................................................................................... 8

2.3 Cell Sourcing for Cardiac Tissue Engineering ................................................................................................... 9 2.3.1 Cardiomyocytes ............................................................................................................................................. 10 2.3.2 Endothelial cells ............................................................................................................................................ 13

Page 6: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

vi

2.3.3 Cardiofibroblast ............................................................................................................................................. 17

2.4 Biomaterials for Cardiac Tissue Engineering ................................................................................................... 18 2.4.1 Properties and Functions of Cardiac Extracellular Matrix ............................................................................ 18 2.4.2 Collagen ......................................................................................................................................................... 19 2.4.3 Elastin and Other ECM Proteins ................................................................................................................... 21 2.4.4 Synthetic Scaffold and Materials ................................................................................................................... 22

2.5 Fabrication and Culture Platforms for Cardiac Tissue Engineering ............................................................. 23

2.6 Angiogenesis and Cardiac Tissue Vascularization Strategies ......................................................................... 25

2.7 In vivo models for evaluating engineered tissue ............................................................................................... 28 2.7.1 Allogeneic and immunosuppressed animal model ........................................................................................ 29

CHAPTER 3: A MODULAR APPROACH TO CARDIAC TISSUE ENGINEERING .... 49

3.1 Abstract ................................................................................................................................................................ 49

3.2 Introduction ......................................................................................................................................................... 50

3.3 Materials and Methods ....................................................................................................................................... 52 3.3.1 Cardiomyocyte isolation and module fabrication .......................................................................................... 52 3.3.2 Endothelial cell seeding ................................................................................................................................. 53 3.3.3 Module sheet .................................................................................................................................................. 54 3.3.4 Electrical response assessment and field stimulation .................................................................................... 54 3.3.5 Immunofluorescence staining ........................................................................................................................ 55 3.3.6 Statistical analysis .......................................................................................................................................... 56

3.4 Results ................................................................................................................................................................... 57 3.4.1 Contractility of CM embedded modules ....................................................................................................... 57 3.4.2 Behavior of RAEC seeded CM embedded modules ..................................................................................... 62 3.4.3 Module sheets ................................................................................................................................................ 64

3.5 Discussion ............................................................................................................................................................. 70 3.5.1 Electrical characterization ............................................................................................................................. 70 3.5.2 Co-culture model ........................................................................................................................................... 71 3.5.3 Sheet formation .............................................................................................................................................. 72

3.6 Conclusion ............................................................................................................................................................ 74

CHAPTER 4: FATE OF MODULAR CARDIAC CONSTRUCTS IN VIVO ................... 79

Page 7: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

vii

4.1 Abstract ................................................................................................................................................................ 79

4.2 Introduction ......................................................................................................................................................... 80

4.3 Materials and methods ........................................................................................................................................ 81 4.3.1 Isolation of rat primary CM and EC .............................................................................................................. 81 4.3.2 Modular cardiac patch model ........................................................................................................................ 81 4.3.3 Peri-infarct module injection model .............................................................................................................. 82 4.3.4 Immunostaining and Histological Analysis ................................................................................................... 83 4.3.5 Statistical analysis .......................................................................................................................................... 84

4.4 Results ................................................................................................................................................................... 85 4.4.1 Characterization of CM embedded modules and rat heart EC ...................................................................... 85 4.4.2 Modular patch and injection implants ........................................................................................................... 88 4.4.3 Host immune response and the removal of MatrigelTM ................................................................................. 97

4.5 Discussion ........................................................................................................................................................... 103 4.5.1 Implantation strategies ................................................................................................................................. 103 4.5.2 Fates of implanted cells and host immune response .................................................................................... 105

4.6 Conclusion .......................................................................................................................................................... 107

CHAPTER 5: FUTURE PERSPECTIVES AND CONCLUSIONS .............................. 112

5.1 Conclusions ........................................................................................................................................................ 112

5.2 Recommendations ............................................................................................................................................. 114 5.2.1 Improving embedded cell viability .............................................................................................................. 114 5.2.2 Using autologous cell source ....................................................................................................................... 116 5.2.3 Characterization of host-module coupling .................................................................................................. 117 5.2.4 Assessing the functional benefits of modular cardiac tissue ....................................................................... 119

Page 8: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

viii

List of Figures Figure 1-1: Schematic of modular construct and modular sheet………………………………...5 Figure 2-1: A schematic diagram showing the major components in the coagulation cascade…………………………………………………………………………………………..15 Figure 2-2: Illustration of components found in endothelial cell tight junctions and adherens junctions…………………………………………………………………………………..……..16 Figure 2-3: Schematic diagram of the three major cardiac tissue engineering strategies……………………………………………………………………...…………………25 Figure 2-4: Changes in the endothelium during angiogenesis…………………………………27 Figure 3-1: Viability (live/dead assay) of cardiomyocytes embedded in modules…………….58 Figure 3-2: Fractional area change of a single module when stimulated by external signal…………………………………………………………………………………………….59 Figure 3-3: Electrophysiological differences between BS and FBS cultured modules……………………………………………………………………………………….…60 Figure 3-4: Electrophysiological differences between modules with and without MatrigelTM……………………………………………………………………………………….61 Figure 3-5: Electrophysiological characteristic of CM/EC co-culture modules……………….63 Figure 3-6A: Schematic diagram of the testing chamber used to condition module sheets……66 Figure 3-6(B-E): Characterization of CM/EC co-culture modular sheets………………… …..67 Figure 3-7: Troponin I expression in cardiac modular sheets………………………………….68 Figure 3-8: Troponin I, vimentin and Cx-43 expression in cardiac modular sheets…………...69 Figure 4-1: Immunofluorescence staining of CM embedded in small diameter modules supplemented with MatrigelTM………………………………………………………………….86 Figure 4-2: Immunoflorescence micrograph of rat cardiac EC isolated from transgenic GFP+ Lewis rats………………………………………………………………………………………..87 Figure 4-3: VE-cadhering expression in EC coated modules before and after injection through a 28G needle………………………………………………………………………………………88

Page 9: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

ix

Figure 4-4: Schematic diagrams and tissue morphology at explant for modular patch implant module injection implant………………………………………………………………………..90 Figure 4-5: Masson’s trichrome stain of patch implants ……………………………….……...91 Figure 4-6: Masson’s trichrome stain of module injection implants…………………………...92 Figure 4-7: Immunohistochemical staining of GFP+ donor cells in explants…………………..93 Figure 4-8: GFP and CD31 expression in patch implants...........................................................94 Figure 4-9: GFP and CD31 expression in module injection implants.........................................95 Figure 4-10: GFP and MHC expression in patch and module injection implants……………………………………………………………………………………….....96 Figure 4-11: Immunohistochemical staining of CD68+ macrophages in patch and module injection implants………………………………………………………………………………..98 Figure 4-12: Immunohistochemical staining of T cells in patch and module injection implants……………………………………………………………………………………….....99 Figure 4-13: GFP and CD31 expression in MatrigelTM-free module injection implants………………………………………………………………………………………...100 Figure 4-14: GFP and MHC expression in MatrigelTM-free module injection implants………………………………………………………………………………………...101 Figure 4-15: Vessel density count in patch and modules injection implants…………………102

Page 10: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

x

List of Tables Table 2-1: Sbtypes of collagen in human ……………………………………………………...20 Table 2-2: A summary of recent implantation studies using various cardiac tissues engineering strategies………………………………………………………………………………………...31 Table 3-1: Medium compositions (presumed key differences) and serum types used for in vitro study……………………………………………………………………………………………..53

Page 11: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

xi

Abbreviations ASC : Adipose stem cell

Ang-1 : Angiopoietin 1

AP : Action potential

BMP-2: Bone morphogenic protein 2

CM : Cardiomyocyte

CF : Cardiofibroblast

Cx43: Connexin 43

d-CMP : Dynamic cardiomyoplasty

EC : Endothelial cell

ECM : Extracellular matrix

EPC : Endothelial progenitor cell

ESC : Embryonic stem cell

FN : Fibronectin

b-FGF : Basic fibroblast growth factor

GAG : glycosaminoglycan

GFP : Green fluorescent protein

IPN : Interpenetrating network

iPS: Induced pluripotent stem cell

MHC-β : Myosin heavy chain beta

MMP : Matrix metalloproteinase

MI : Myocardial infarction

bmMSC : Bone marrow derived mesenchymal stem cell

MSC : Mesenchymal stem cell

NO : Nitric oxide

PGA : Polyglycolic acid

PLA : Polylactic acid

PCL : Polycaprolactone

PDGF : Platelet derived growth factor

PDMS : Polydimethylsiloxane

PFA : Platelet activating factor

Page 12: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

xii

SMC : Smooth muscle cell

TFPI : Tissue factor pathway inhibitor

TF : Tissue factor

TGF-β : Transforming growth factor beta

TnI : Troponin I

VEGF : Vascular endothelial growth factor

vWF : von Willebrand factor

Page 13: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

1

Chapter 1: Research Introduction, Hypotheses and Objectives

1.1 The Need for Engineered Cardiac Tissue

The heart is a complex organ with a simple function: to pump blood throughout the body

without interruption over an individual’s lifespan. However, certain diseases including coronary

artery disease and cardiomyopathy can cause cell death in the myocardium. Unlike skin or liver

tissues, adult myocardium has limited regeneration capacity. Thus, any loss of cells usually

translates to a permanent loss of cardiac function. On a tissue level, cell debris draws in

inflammatory cells and contributes to the formation of scar tissue. Ultimately, the combination

of tissue loss and scar tissue formation will further decrease the output of the heart and cause

more damage to the rest of the myocardium.

The vicious cycle of functional loss due to cell death leading to further tissue damage

could be stopped, and possibly reversed, by replacing diseased myocardium with functional

cardiac tissue. Tissue engineering has made it plausible to create tissues in vitro for transplant

and replacement of scar tissue. To date, several promising methods have been devised for

building cardiac tissue in vitro, including cell seeded scaffolds, cell-embedded hydogel, or

matrix-free cell sheets. However, many of these engineered cardiac tissues lack a functional

vasculature to sustain a high cell density. The challenges in creating cardiac tissue with

physiologically relevant thickness and function remain to be overcome before they can achieve

widespread clinical significance.

Page 14: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

2

1.2 Research Overview and Hypothesis

The overall goal of this work was to create functional, implantable cardiac tissue from a

modular tissue engineering construct embedded with neonatal rat cardiomyocytes (CM). We

hypothesized that the modular tissue engineering construct developed in our laboratory1 would

serve as a useful platform for producing vascularized cardiac tissue in vivo. A module is a small

cylindrical collagen gel with a length of approximately 1500 µm and diameter of 500 µm.

Modules were embedded with a CM-rich cell mixture composed of CM, fibroblast, cardiac

progenitor stem cells, and endothelial cells (EC). We coated the surface of each module with a

monolayer of EC with the view that such design would provide the means to control initial cell

seeding density, reduce thrombogenicity and improve scaffold vascularization in vivo. We

examined the responses of CM-only as well as cardiomyocyte-endothelial cell (CM-EC) co-

culture modules to external electrical stimulation. In addition, we assessed the effect of surface

coated EC on the maturation and function of embedded CM.

The fate of the cardiac modular construct in vivo was examined using syngeneic animal

models. Cells from transgeninc eGFP Lewis rats were used to construct modules and then

implanted into host Lewis rats. Cell survival, maturation and immune responses were

monitored. We hypothesized that the presence of EC on CM-EC co-culture modules would

increase implant vascularity and lead to subsequent improvements in overall cell survival and

tissue morphology compared to CM only modules. Also, we expected the cardiac modular

construct, with or without EC, to be well tolerated by their syngeneic host with minimal immune

response.

Page 15: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

3

1.3 Research Objectives

1.3.1 Overall Objectives

The overall objective of this project was to create cardiac tissue using a modular tissue

engineering construct. To achieve this we had to 1) devise a method of fabricating CM-EC co-

culture modules and higher order structures, 2) characterize modular cardiac tissue using

functional in vitro assays, and 3) investigate the fate and remodeling process of cardiac tissues

using in vivo models. These three objectives will be covered in the following section.

1.3.2 Specific Objective 1: Fabrication of CM-EC co-culture modules

Modular co-culture constructs (CM-EC) were fabricated using a collagen based modular

engineering construct developed in our laboratory. Each module was embedded with a CM-rich

cell mixture harvested from neonatal rat heart and the surface was coated with a monolayer of

rat EC (Figure 1-1A).

We have also devised a method to assemble modules into a single, sheet-like structure

with thickness and pores the size of a module (Figure 1-1B). Modules embedded with CM

underwent spontaneous agglomeration and fusion when placed in close proximity to one

another. By immobilizing a single layer of modules in alginate gel, intermodular junctions were

fused together while alginate, a non-cell adherent natural polymer, acted as a physical and

biochemical barrier to prevent the collapse of interstitial spaces between modules and to

maintain sufficient permeability for nutrient transport. An interlinking, perfuseable,

macroporous modular network was obtained by dissolving the alginate gel with a citric acid

buffer.

1.3.3 Specific Objective 2: Characterization of CM/EC co-culture modules

Earlier studies have demonstrated that collagen modules can support and maintain

surface coated HUVEC and embedded HepG2 cells2. However, the functionalities of embedded

Page 16: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

4

cells have not been investigated. In this project, both CM and RAEC were assessed for the

presence of functional phenotypes. Electrical responsiveness and contractility were measured as

primary indicators of embedded CM functionality. While, the expression of cardiac specific

markers, including troponin I, troponin T and myosin heavy chain on immunohistological

sections served as secondary markers. For surface coated EC, VE-cadherin expression was used

as a quiescent EC marker.

1.3.4 Specific Objective 3: Fate of CM/EC co-culture modules in vivo

The interactions between engineered tissues and their hosts can significantly impact the

viability and functionality of the implant. In this project, we examined the fate of the modular

cardiac construct in vivo using a syngeneic rat model. Cells harvested from neonatal eGFP

transgenic Lewis rats were incorporated into modular constructs and implanted into the hearts of

Lewis rats. We used two separate implantation models to determine the best method of

deploying the modular construct into the heart and also to observe their impacts on host-graft

interactions. Our first model involved the construction of a cardiac patch, where macroporous

cardiac modular sheets were sandwiched between two pieces of porous gelatin foam, also know

as GelFoam. These patches were then implanted over a transmural wound on the right

ventricular free wall of a healthy Lewis rat heart and explanted after 2 weeks. The second model

attempted to determine the fate of cardiac modules when implanted as individual microtissues in

the heart. An acute myocardial infarct (MI) was induced in a Lewis rat by ligation of the left

descending coronary artery. After 7 days, small diameter modules embedded with CM were

injected into the peri-infarct zone and explanted after 21 days. Explanted tissues were assessed

for cell viability and morphologies by histological analyses (H&E, Mason Trichrome, eGFP,

TnI, TnT, MHC, CD31, CD68, T-cell receptor, and other relevant immunohistochemical

stainings). Blood vessel density (both GFP positive and GFP negative) were measured as a

surrogate marker for angiogenesis.

In both models, we expected CM-embedded modules implanted into the rat heart to

develop into more native-like morphology compared to CM-modules in vitro. Also, we expected

the presence of EC in co-culture modules to improve overall cell survival and illicit a more

pronounce angiogenic response. Finally, in the injection model, we expected the modular

Page 17: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

5

approach to yield better cell engraftment and improve survivability compared to conventional,

matrix-free cell injection therapy.

Figure 1-1: Schematic diagrams of a CM embedded EC coated module (A) and a single layer,

porous modular sheet (B). Modular sheet were fabricated by immobilizing a single layer of

modules on a nylon mesh with alginate gel. After the intermodular junctions have fused, the

alginate was removed using a citrate buffer wash, leaving behind a porous modular sheet.

Details about the fabrication and characterization of modular cardiac construct will be covered

in Chapter 3 and 4.

1.4 Thesis Organization

The thesis is organized into self contained chapters. The first chapter outlines the scope and

objectives of the thesis. While, chapter 2 goes on to review and present the current methods and

challenges in cardiac tissue engineering as well as relevant background literature. In chapter 3

and 4, we discuss the fabrication of modules and their behavior in vitro. Specifically, chapter 4

focuses on the fate of modular constructs in vivo. In the last chapter, we summarize the lessons

learned from the project and evaluate the usefulness of the modular construct in the field of

cardiac tissue engineering. We also discuss the future direction of the project in chapter 5.

Page 18: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

6

Reference List

1. A. P. McGuigan and M. V. Sefton, Design criteria for a modular tissue-engineered

construct. Tissue Eng 13, 1079-1089 (2007).

2. A. P. McGuigan and M. V. Sefton, Vascularized organoid engineered by modular

assembly enables blood perfusion. Proc. Natl. Acad. Sci. U. S. A 103, 11461-11466

(2006).

Page 19: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

7

Chapter 2: Current Methods and Challenges in Cardiac Tissue Engineering

2.1 Engineered Cardiac Muscles for Functional Cardiac Regeneration

Similar to other organs, the heart owes its specialized functions to the unique complement

of cells and their complex arrangements. Its contractile function derives primarily from

cardiomyocytes (CM). These myocytes are tightly packed with neighboring fibroblasts, which

act as supporting cells by maintaining the extracellular matrix (ECM) as well as guiding

electrical current propagations throughout the heart. On the other hand, the myocytes’ demand

for oxygen and nutrients are satisfied by a dense endothelial cell (EC) lined vasculature.

Together these three cell types form a mechanically and electrically coupled unit that generates

the force necessary to pump blood throughout the body.

Until the recent discovery of resident cardiac progenitor cells1,2, the myocardium was

thought to be composed entirely of terminally differentiated cells. However, in their native state,

these progenitor cells do not contribute sufficiently to the repairing of major cardiac trauma.

Thus, from a clinical perspective, the adult heart has little capacity for regeneration after injury.

A constant supply of oxygen and nutrient is required to sustain the high metabolic demand of

CM. For this reason, any disruption of circulation to the heart muscle, such as that seen during a

myocardial infarction (MI), will lead to rapid cell death and tissue necrosis. In most cases, the

cell debris triggers an inflammatory response that is characterized by the infiltration of

monocytes and macrophages. The necrotic tissue is then replaced by granulation tissue

consisting mainly of fibroblasts and collagen. Finally, this tissue is remodeled to form a stiff and

non-contractile scar tissue.

Recent studies have demonstrated that injection of cells into the myocardium, including

bone marrow derived mesenchymal stem cells (bmMSC)3,4, skeletal myoblasts5-7 and CM

derived from embryonic stem cells8,9, may prevent scar tissue formation and restore cardiac

function in patients with acute MI. It has also been reported that circulating endothelial

progenitor cells (EPC) from the patients’ bone marrow may become mobilized and participate in

angiogenesis in the peri-infarct zone10. A comprehensive review of current theories on cell-

Page 20: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

8

based therapies for MI has been published by Laflamme et al.11 and hence will not be covered

within this chapter.

In many cases, the presence of scar tissue and extensive tissue remodeling caused by

chronic ischemic cardiomyopathy requires the complete replacement of damaged tissue with

functional ones in order to achieve significant clinical benefits. Early attempts such as dynamic

cardiomyoplasty (d-CMP) aimed to replace cardiac function by implanting an autologous

skeletal muscle flap taken from the patients’ body. Although the implanted muscle flap

improved cardiac performance, the progressive degeneration of the implant and the lack of

electrical coupling with host tissue limited its clinical usefulness12. While d-CMP failed as a

replacement therapy to prevent total heart transplantation, it sparked the notion that one could

fabricate implantable cardiac tissue in vitro by combining CM and other supporting cell types in

an appropriate scaffold. The resulting engineered cardiac tissue should possess similar

mechanical and functional properties as native cardiac tissue. The presence of functional cells

should have the capability to secrete ECM and growth factors to encourage beneficial host

remodeling, which will significantly contribute to the long term integration of the implant13.

2.2 General Considerations for in vitro Tissue Engineering

In vitro engineered tissues could provide the means to rapidly replace tissue functions that

are lost due to trauma or acute diseases. These tissues may be biomimetic in nature, or they

could be designed to functionally imitate the target tissue. However, there are many obstacles

facing the clinical implementation of these therapies. Strategies for engineering tissue in vitro

must address the following elements:

1. Providing a mechanically and biochemically suitable extracellular environment to support

cellular function.

2. Finding the appropriate cell source and/or incorporating bimolecular cues into the scaffold

to promote recruitment of functional endogenous cells.

3. Providing sufficient perfusion of oxygen and nutrients to embedded cells.

4. Providing the necessary environmental stimuli to promote proper tissue development.

5. Integrating engineered tissue structurally and functionally with the host tissue.

Page 21: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

9

The biological and structural functions of engineered tissue are intimately related to the

phenotypes of the incorporated cells. Besides the obvious differences among cell types, it is also

important to consider the subtle phenotypic differences of cells from different origins and

lineages. For example, while most EC in various organs share common EC markers, there are

heterogeneous subpopulations of EC within the endothelium and even within the organ.

Functional differences between EC found in the brain, kidney, liver and gastrointestinal tract

manifest as different EC phenotypes. Mechanical forces in large diameter vessels versus those

associated with capillary blood flow contributes to functional and morphological differentiation

of EC14. Moreover, paracrine signaling by cytokines often causes local differential EC gene

expression15.

While achieving the necessary mechanical properties are essential, the design for

engineered tissues must also satisfy the basic requirements for supporting cellular growth.

Among the most crucial criterion is the need to provide adequate mass transport to and from the

implanted cells so that nutrients and waste products can be exchanged freely. Under desirable

circumstances, adequate blood supply into the tissue also ensures that the secreted therapeutic

molecules from the tissue can reach the systemic circulation.

Beyond the basic survival requirements of nutrient delivery, cells also need a suitable

extracellular environment to engage in cellular functions such as proliferation, migration and

differentiation. Selection of a tissue engineering scaffold that resembles in vivo ECM

architecture not only enhances the functionality of embedded cells, but may also encourage the

migration of endogenous cells into the implant and peri-implant region. Natural ECM

components possess many of the desirable qualities as a building block for tissue engineering

scaffolds. Considerations of cell sourcing, vascularization and ECM interaction in the modular

tissue engineering scaffold are discussed in detail within the following sections.

2.3 Cell Sourcing for Cardiac Tissue Engineering

Cell sourcing is perhaps the most important consideration when designing tissue

regeneration strategies. The choice of cell can directly impact host response, ECM remodeling,

and ultimately the functioning of the regenerated tissue. The cells selected should also be

matched with suitable scaffolds, culture conditions and delivery methods in order to maximize

its therapeutic potential. In cardiac tissue engineering, the cells of interest are primarily

Page 22: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

10

cardiomyocytes, cardiofibroblasts, and endothelial cells. The following sections will outline the

features of these cell types and their most common sources.

2.3.1 Cardiomyocytes

The most striking feature of the heart is its ability to contract continuously and

synchronously during a person’s lifespan. Heart muscle cells, called cardiomyocytes (CM), are

the primary cell type responsible for the generation of contractile force. The developmental

origin of CM can be traced back to the mesoderm layer of the gastrulated embryo16. The heart

itself is formed by two separate waves of myogenesis known as the primary and secondary heart

fields. The primary heart field originates from the anterior lateral mesoderm and give rise to the

left ventricle of the mature heart, while the secondary heart field develops into the right ventricle

inflow and out flow tracks17. The secondary heart field also give rise to some of the non-

myocyte component of the heart including cardiofibroblast (CF), smooth muscle cells (SMC)

and endothelial cells (EC).

CM share many similarities with skeletal muscle cells; both are striated, multinucleated,

and excitable cells. In comparison to skeletal muscle cells, CM are more resistant to fatigue,

contain more mitochondria, and are more adapted to aerobic respiration. Nevertheless, perhaps

the most distinct anatomical feature of CM are the intercalated discs found between adjacent

cells. Intercalated discs are areas of cell membrane rich in desmosomes and gap junctions.

Desmosomes are cadherin family transmembrane proteins that are associated with intermediate

filaments. Through homophilic binding, desmosomes also confer mechanical cell adhesion and

integrity between CM. On the other hand, intercellular communications are mediated by gap

junctions. Transmembrane proteins called connexins form hemi-channels on the membranes of

adjacent cells. These connexons can align and form a direct channel between the cytoplasm of

these adjacent cells, allowing membrane depolarization to propagate from one cell to the next,

thus facilitating action potential propagation over the myocardium.

The ultimate goal of cell sourcing in tissue engineering is to find a cell type that can

differentiate into all the necessary cells found in an organ in order to recapitulate organogenesis

during development. Perhaps, the cells that carry the most potential toward this goal are

embryonic stem cells (ESC) derived from the inner cell mass of the blastocyst. These cells are

Page 23: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

11

pluripotent and have strong proliferative capacity compared to adult stem cells. However,

pluripotency is not without its downfalls. Direct injection of undifferentiated ESC can lead to

teratoma formation18. Therefore, the phenotype of ESC and ESC derived cells must be carefully

regulated. Selective differentiation of ESC have been shown to be a promising path towards a

steady source of CM19-21. Laflamme et al. demonstrated in a rat model that the injection of

differentiated human ESC maintains cardiac phenotypes and continues to proliferate over 4

weeks22. In addition, the same group has demonstrated the utility of human ES derived CM in

tissue engineering application by fabricating contractile cardiac constructs23.

Although ESC derived CM improved cardiac function in vivo, their allogeneic nature

poses a significant obstacle towards clinical application. The recent discovery of induced

pluripotent stem (iPS) cells derived from adult fibroblast may be the answer for an autologous

and pluripotent cell source. The Yamanaka group pioneered the technique in restoring

pluripotency of somatic cells to the same level as ESC by ectopically expressing Oct3/4, Sox2,

Klf4 and c-Myc24. It can be assumed that current methods being used to drive ESC toward CM

differentiation can similarly be applied to iPS cells. Like other stem cells, the in vivo

tumorgenicity and immune response toward iPS cells will ultimately determine their clinical

usefulness. Further studies are needed to answer these questions.

From a clinical therapy perspective, the ideal CM for tissue engineering should retain

contractile phenotype indefinitely, cause minimal immune response in the host, and participate

in functional remodeling by organizing into contractile bundles25. Much of these criteria can be

satisfied by using CM differentiated from stem or progenitor cells. Studies in stem cell lineages

have identified populations of adult stem cells in tissues, such as adipose26, skin, dental pulp27

and amniotic fluid28,29, some of which are capable of differentiating into beating CM30.

However, the most studied population for cardiac regeneration in clinical application to date is

the bone marrow derived messenchymal stem cells (MSC). These cells inhabit the stromal space

of bone marrow. They can be obtained through direct bone marrow aspiration or from peripheral

blood. In both cases, the MSC reside as a subpopulation of cells within the mononuclear cell

fraction31. Under the right condition, these MSC can be induced to express cardiomyogenic

markers32 and play a cardioprotective role to nearby cardiomyoblasts33. In clinical studies,

intravenous or intramyocardial injections of MSC in acute MI have been shown to increase

ejection fractions and vascularity around the infarct area34,35. However, few MSC differentiate

into CM lineage and those that are committed have little to do with the functional improvement

Page 24: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

12

that are observed36. Therefore, it appears that much of the benefits of MSC in acute MI are

derived from their cytoprotective and angiogenic roles37,38.

Another potential source of autologous CM can be found in adipose-derived stem cells

(ASC). Like MSC, these cells can be kept undifferentiated over long periods of time in culture

and they are capable of differentiating into muscle, bone and fat cells in vitro. Even though the

immunophenotype of ASC closely resemble that of MSC39, they behave very differently in

culture. ASC are capable of differentiating spontaneously into beating CM through vascular

endothelial growth factor (VEGF) paracrine signalling40. A similar phenomenon can also be

observed when ASC are cultured with cell extract of neonatal rat CM41, with TGF-β alone30, or

in the presence of explanted cardiac tissue42. Moreover, the induction of ASC into myogenic

phenotype by surrounding CM can be observed in vivo when ASC are injected intracardially in

an acute MI model43. Due to their strong VEGF paracrine signaling, ASC can promote

angiogenesis and stimulate vessel growth when cultured with neonatal hearts containing CM

and EC44. Intramyocardially injected ASC were also shown to significantly improved cardiac

function after MI in animal models45,46. These results have demonstrated the therapeutic

potential of ASC in cell therapy as well as a source of autologous CM for cardiac tissue

engineering.

Although not a viable CM source for therapeutic applications, primary rat neonatal

ventricular cardiomyocytes have served as a useful cell model that has greatly contributed to the

understandings of CM physiology and development. These CM differ from adult ventricular

CM in its ability to grow and differentiate during the transitional phase from neonatal to adult

myocytes in culture47. Structurally, neonatal CM lack a well developed T-tubule system, the

deep invagination of the sarcolemma, found in adult CM48. This leads to a more heterogeneous

cytosolic Ca2+ concentration compared to adult myocytes and contributes to its unique

excitation-contraction coupling characteristic.

Page 25: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

13

2.3.2 Endothelial cells

Endothelial cells (EC) are found on the luminal surface of all blood carrying vessels. A

monolayer of EC, known as the endothelium, has been accepted as a separate organ weighting 1

kg in an average human that lines the entire circulatory system49. The lineage of EC is closely

related to hematopoietic cells. It is known that both cells differentiate from a common precursor

referred to as the hemangioblast. Although EC are considered a terminally differentiated cell

type, they are also believed to transdifferentiate into intimal smooth muscle cells (SMC)50-52.

Physiologically, EC are rarely present without cellular interaction with supporting cell types

such as pericytes and SMC. In a large diameter vessel, a layer of intimal SMC underlies the

endothelium, and together controls vascular tone by various paracrine and autocrine pathways.

At the capillary level, the endothelium is associated with pericytes, SMC-like cells. The

endothelium has long been considered as an inert barrier between blood and the underlying

tissues. However, recent advances in basic EC biology and cardiovascular research have

identified numerous biological and metabolic processes involving the endothelium. The

endothelium serves as a haemostatic organ by dynamically balancing pro-thrombotic and anti-

thrombotic signals. It is actively involved in controlling transendothelial transport of molecules,

gating the migration of various immune cells from circulating blood into interstitial space, and

may act with or against inflammatory responses. The endothelium is also an integral part of

establishing and maintaining vascular tone by sensing and responding to changes in shear stress

induced by blood flow. From a tissue engineering perspective, EC provide the building block for

enabling functional blood vessel to carry nutrients into the seeded cells. Consequently, the

following sections will cover the attributes of EC that are of primary interest, namely their

haemostatic and angiogenic (Ch 2.6) properties.

It is the haemostatic property of the EC that makes it an important component in a tissue

engineering scaffold vascularization strategy. The endothelium actively regulates thrombosis

and thrombolytic factors in response to platelet adhesion, blood chemistry, vasomotor tone and

blood flow regime53. The endothelium can be described in one of two states: (1) a basal or

quiescent state, where EC are non-proliferative and display low platelet adhesion, or (2) an

activated state, where EC phenotype change to a pro-thrombotic, proliferative state. The

endothelium can be triggered to switch from a quiescent to an activated phenotype by

pathogens, injury signals (tissue factor, collagen), or chemical cues (thrombin).

Page 26: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

14

The basal state of the endothelium actively suppresses intravascular thrombosis by

secreting tissue factor pathway inhibitor (TFPI), which binds with factor Xa and tissue factor-

VIIa to form an inactive complex54. While TFPI is primarily bound to the EC membrane, it may

also be released into the circulation in a soluble form. Factors such as heparin, thrombin and

blood flow increases the production and/or release of TFPI55-57. EC also secrete

glycosaminoglycans (GAGs), namely heparan sulfate, that catalyze the inhibition of factor Xa

and thrombin by antithrombin58. Moreover, basal state endothelium mediates protein C (PC)

activation and inhibits thrombosis by expressing thrombomodulin (TM). TM is a membrane

bound protein that binds thrombin. The formation of TM-thrombin complex has two

anticoagulation effects. First, it prevents thrombin from activating fibrinogen, platelets, factor V,

VIII and XIII59. Secondly, the TM-thrombin complex activates protein C via the endothelial

protein C receptor (EPCR), which inactivates factor Va and VIIa and disables the synthesis of

thrombin60.

One of the major procoagulation proteins that are expressed by EC is tissue factor (TF).

Although TF is known to be constitutively expressed by subendothelial vascular cells, it is only

expressed by EC in an activated state61. TF forms a complex with plasma serine protease factor

VII/VIIa and in turn activates IX and X to initiate the blood coagulation cascade. In quiescent

endothelium, TF transcription is actively suppressed62. The endothelium can be activated by

many agonists including vascular injury, alteration of blood flow regime, and infection. Bacteria

derived endotoxins have been used routinely to activate EC in culture. In its activated state, EC

also express platelet activating factor (PAF), which together with P-selectin causes the adhesion

of platelet to the endothelium49. Platelet adhesion is often viewed as a critical step in the

initiation of intravascular thrombosis. The activated endothelium may also facilitate thrombosis

by releasing von Willebrand factor (vWF) to act as a stabilizer for factor VIII and promote its

binding to ECM components.

Page 27: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

15

Figure 2-1: A schematic diagram showing the major components in the coagulation cascade.

The cascade is based on the sequential conversion of clotting factor precursors into active

proteases. The ultimate result is the generation of a fibrin, which then polymerizes and

crosslinks to form a clot. (Adapted from NCBI tutorial: Mutation and blood clot, CC)

The endothelium is one of a handful of non-structural organs that is capable of sensing

and responding to mechanical changes in its environment. EC from different regions of the

circulatory system are known to have different responses to external stimuli, including their

sensitivity toward hemodynamic forces63-65. Platelet-endothelial cell adhesion molecule

(PECAM or CD31) is an EC specific transmembrane adhesion protein that plays a major role in

mechanosignal transduction. The extracellular domain of PECAM participates in cell-cell

adhesion through homophilic association while the intracellular domain is associated with

various adaptor proteins through tyrosine phosphorylation and is ultimately associated to the

cytoskeleton. Integrins are another class of transmembrane proteins that are responsible for

mechanosignal transduction. Conformational activation of integrins induced by fluid shear is

responsible for adaptive EC morphogenesis including cytoskeletal alignment. However, the

Page 28: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

16

extracellular domains of integrin are mainly associated with ECM components and hence they

are not considered as a mechnosensory components to fluid induced shear stress66,67.

As an organ that controls mass transport across the blood-tissue barrier, it is imperative for

EC to have strong and tight intercellular adhesion. Junctional components found between EC are

associated with one of the two specialized regions on the plasma membrane, namely tight

junctions and adherens junctions. Each type of junction contains its own unique membrane

proteins (Figure 2-2).

Figure 2-2: Illustration of components found in endothelial cell tight junctions and adherens

junctions68.

Tight junctions are located on the apical (or luminal) side of the EC cell-cell junction.

Their main function is to seal off the space between EC to give a continuous luminal surface in

order to facilitate blood flow. The main proteins involved in tight junction architecture are

occludins, claudins, and junction adhesion molecules (JAM)69. Their proximity to the lumen

allows them to serve as mechnosignaling sensors70.

Unlike tight junctions, adherens junctions are responsible for the mechanical integrity of

cell-cell junctions and intercellular signaling. A family of cell specific proteins known as

cadherins maintains adherens junctions. In EC, two types of cadherin are expressed: vascular-

endothelial cadherin (VE-cadherin or CD144) that localize to adherens junctions only and N-

Page 29: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

17

cadherin that are found dispersed over the plasma membrane69. The function and structure of

VE-cadherin are well characterized and it is believed to be the major component of adherens

junctions. The extracellular domain of VE-cadherin exhibits homophilic, calcium dependent

associations. On the other hand, its intracellular domain associates with plakoglobin and β-

catenin, which ultimately connects to the actin cytoskeleton71. Similar to PECAM, the

intracellular domain of VE-cadherin can undergo tyrosine phosphorylation, which has been

shown to destabilize adherens juctions72 and may be used as a pathway to modulate endothelium

permeability. For this reason, the expression and localization of VE-cadherin has been used as a

vessel maturation and endothelium stability marker73. One of the receptors for VEGF, VEGFR-

2, mediates VE-cadherin tyrosine phosphorylation upon VEGF binding and is believed to

potentiate the sprouting response in early stage of angiogenesis74. The importance of VE-

cadherin as a structural component in the adherens junctions is further substantiated by attempts

to develop VE-cadherin knockout mice, which was demonstrated to result in embryonic

lethalaty75 due to impaired vessel development.

2.3.3 Cardiofibroblast

Cardiofibroblast (CF) of messenchymal origin constitutes the bulk of non-myocyte cell

population in the heart76. The main function of CF is to secrete and maintain ECM components,

including collagen, fibronectin, and laminin. Unlike CM and EC, CF is found natively in the

stromal space and lack basement membranes. This feature gives CF its unique ability to migrate

and populate injury sites, such as a myocardial infarct, and quickly restore tissue volume and

ECM proteins77. In the developing fetal heart, CF contributes to ECM rich structure including

valves and atrial ventricular walls. As the heart matures, a 3D network of collagen and CF,

known as the cardiac skeleton, begins to take shape. This network allows CF to exert forces on

the myocytes, as well as, to respond to external stimuli through degradation and synthesis of

ECM. These processes help maintain the mechanical integrity of the heart through cell-cell and

cell-ECM interactions. More importantly, CF forms heterotypic gap junctions with neighboring

CM or CF, whereby the conductivity of these junctions can be modulated by the differential

expression and coupling of connexins isoforms, including Cx45, Cx43 and Cx4078,79. The cell-

Page 30: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

18

cell interaction between CF and CM helps to ensure the long range synchronization of

myocardium contraction.

CF also plays a pivotal role during cardiac repair. Residence CF can mobilize in

response to tissue damage and differentiate into smooth muscle-like cells called myofibroblast80.

These are not true smooth muscle cell, but they are more contractile and active compared to

fibroblast. The ability of myofibroblast to contract collagen and produce new ECM helps

stabilize wound site and form scar tissue. In myocardial infarcts, these cells are responsible for

the formation of granulation tissue. However, their eventual loss through apoptosis results in the

weakening of the scar and the ultimately leads to heart wall thinning77,81. Therefore, the dynamic

balance between fibroblast and myofibroblast phenotype is a critical factor in the wound healing

outcome.

2.4 Biomaterials for Cardiac Tissue Engineering

2.4.1 Properties and Functions of Cardiac Extracellular Matrix

Extracellular matrix (ECM) refers to the non-cellular components found in tissue. It is a

complex mixture of molecules including proteins such as collagen, elastin, laminin, fibronectin

and polysaccharides such as glycosaminoglycan (GAG). The composition and architecture of

ECM varies depending on the native tissue. The primary purpose of ECM is to provide

structural integrity to tissue. However, it is now known that ECM components such as collagen

and laminin also play a crucial role in functional cell signaling through ECM receptors, such as

the integrins, on cell membranes82-85. ECM of load-bearing tissues such as tendons and bones

may persist for long periods of time and have a much lower turnover rate compared to ECM in

skin and mucosal membrane, which are constantly degraded and re-synthesized to accommodate

cellular functions such as cell migration and proliferation. The majority of ECM components in

the heart are secreted by cardiofibroblasts and myofibroblasts. They provide a wide range of

ECM molecules for various functions, including fibrillar collagen and elastin for structural

integrity, laminin and collagen IV for basement membrane maintenance, and proteoglycans for

cell signaling modulation86,87. Moreover, these ECM components are able to transmit external

stimuli to the myocytes and fibroblast and trigger intracellular signaling that can alter cellular

Page 31: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

19

functions. Recent work by Ott et al. demonstrated that decellularized native cardiac tissue can

be repopulated with CM and CF and give rise to functional tissue that possesses the

microarchitecture of native tissue88. This suggests that both the composition and organization of

ECM molecules are important for proper tissue development. The following section will cover

the properties of these major ECM molecules.

2.4.2 Collagen

It is the most abundant protein in the body and accounts for 20 to 30% of total body

weight89. Collagen has a simple amino acid sequence consisting of tripeptide repeats. This leads

to a high degree of homology among collagens from different species. For example, type I

collagen from bovine and porcine skin has been demonstrated to be highly compatible with

human cells90. The abundance and compatibility of collagen makes it an ideal matrix substrate

for tissue engineering applications.

The widespread presence of collagen in the ECM is attributed to its unique micro and

macro structure. Collagen is secreted mainly by fibroblasts, smooth muscle cells and EC, and it

is present only in the ECM. The basic primary structure of collagen consists of repeating tri-

peptide units of -Gly-X-Y-, where the amino acid X and Y are often represented by proline and

hydroxyproline90 respectively. The space and flexibility provided by the small hydrogen residue

on glycine is essential for the assembly of collagen polypeptide into more complex, superhelical

secondary structures, while the ring structure of proline stabilizes the superhelix.

Each collagen polypeptide chain forms an extended left-handed helix. Three helices self

assemble to form a trimeric right-handed superhelix91. The triple helix is held together through

hydrogen bonds among the three associated chains. These triple chain molecules comprise the

basic units for larger collagen structures, such as collagen fibrils, which are parallel, staggered

bundles of collagen superhelices. Polypeptides of different collagen isoforms, or α-chain, may

also associate to form heterotrimers; thus, yielding a large variety of collagen fibrils of different

mechanical properties. More than 20 different collagens α-chains have been identified and each

has their unique primary and secondary protein structure. Based on amino acid sequence,

microstructure and functional attributes, collagen can be subdivided into several types. Type I

collagen is the most abundant collagen. Togethertype I, II, III and V collagen share the same

triple-helix structure and are referred to as fibrous forming collagens. In cardiac tissue, over

Page 32: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

20

90% of collagen belongs to either type I or III92. Other types of short fibrous collagen (type IX,

XI, XII and XIV) serve as anchor fibers to secure cells and other ECM components to the larger,

continuous collagen fibers. Type IV collagen is found in basement membrane and has unique

globular structures along the triple-helix backbone93 resulting in the formation of sheet-like

structure. Common collagen subtypes and their tissue distributions are listed in Table 2-1.

Table 2-1: Subtypes of collagen in human (adapted from Table 19-5, Molecular Biology of Cell,

4th edition).

There are many characteristics that make collagen a desirable material for tissue

engineering scaffolds. Collagen is an excellent substrate for cell attachment. Certain amino acid

sequences found in collagen are targets for integrin receptor binding. For example, native type I

collagen expresses the tetrapeptide domain -Arg-Gyl-Asp-Ala- (RGDA), which binds the

ubiquitously expressed α1β1 and α1β2 integrin66.

Collagen can be readily remodeled by matrix metalloproteinase (MMP) secreted from

implanted cells or native tissues surrounding the implant. Various experimental and

Page 33: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

21

commercially available collagen products such as IntergraTM (Intergra Lifesciences Inc.) are

designed to gradually degrade and be absorbed by peri-implant tissues. As a natural matrix

component in bone, collagen and collagen-elastin matrix with embedded cytokines such as bone

morphogenic protein-2 (BMP-2) have been used as a osteogenic matrix for bone regeneration94-

96. Unfortunately, collagen is a natural agonist for thrombosis via platelet activation97.

Therefore, it is a poor choice for blood contacting applications.

In the adult myocardium, the five most common types of collagen, in descending order

of abundance, are type I, III, IV, V and VI. Collagen plays several essential roles in cardiac

ECM. It mediates the fibrotic response of CF by forming complexes with nonadhesive signaling

protein, such as osteopontin, through the integrin receptors98. Type I collagen is the major

component of the perimysium, a sheath-like connective tissue which surrounds and

interconnects groups of myocytes to help maintain the ordered laminar structure of muscle

bundles in the myocardium99,100. Each laminar layer consists of several fiber bundles stacked

together. The layers are organized in varying directions depending on their trasmural locations.

Fibers located on the epicardial and endocardial surfaces run in a base-apex orientation, whereas

fibers in the mid-wall run in a circumferential orientation101. This structure gives rise to the

anisotropy of the myocardium and also magnifies the shorting of each fiber bundle into gross

ventricular wall thickening that is much greater than anticipated102.

From a biomaterial perspective, collagen is a highly versatile material. It can be easily

extracted from a wide variety of sources and can be processed into many different forms from

sheets to discs, and hydrogel sponge to electrospun mesh. The amino acid residues on the

collagen polypeptide backbone can be readily crosslinked or modified with synthetic polymers

to modulate its mechanical and biological properties. For example, an interpenetrating network

(IPN) made from poloxamine, a synthetic hydrogel, and natural collagen fiber has been shown

to improve the mechanical strength of the resulting composite hydrogel while maintaining

cytocompatability103.

2.4.3 Elastin and Other ECM Proteins

Elastin fibers provide resilience and elasticity to tissues such as blood vessels, skin and

cardiac tissue. In arteries, elastin fibers constitute up to 50% of its dry weight and is secreted

Page 34: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

22

mainly by vascular smooth muscle cells104. Moreover, elastin fibers are long-lasting and have a

low turnover rate, thus making it an important structural ECM component.

The basic molecular unit of elastic fibers is elastin. Like collagen, elastin is synthesized

in a soluble pro-protein form known as tropoelsatin. Tropoelastin is bound by a 67 kD

galatolectin chaperone protein on the cell membrane to prevent premature coacervation105.

Tropoelastin consist of alternating Lys-Ala rich hydrophilic and Val-Pro-Gly rich hydrophobic

domains106. Unlike collagen, no proteolytic processing of tropoelstin is required for

elastogenesis. Instead, the lysine/alanine rich domain takes on the α-helix structure, where the

lysyl residues are readily deaminated by extracellular lysyl oxidase to form translysyl

crosslinkages107. On the other hand, hydrophobic domains from adjacent tropoelastin molecules

form loose interpenetrating chain structures that are responsible for the elasticity of elastin

fibers108. Once crosslinked, elastin forms an insoluble elastin fiber.

Besides collagen and elastin, fibronectin (FN) is the next most abundant ECM

component. FN molecules form homodimers and consist of several distinctive functional

domains. FN exists in both insoluble and soluble forms. In its insoluble ECM form, FN

associates with collagen fibers with its collagen-binding domain, while interacting with cell

surface adhesion molecules such as integrin through its Arg-Gly-Asp (RGD) domain. This

bifunctional nature allows FN to serve as a biological crosslinker between cells and the ECM109.

Also, by using integrin as a transmembrane adapter protein, the actin cytoskelekton exerts forces

on extracellular FN fibrils to aid in its polymerization and organization104. Soluble FN circulates

through the blood stream and participates in thrombosis and wound healing.

Another bifunctional ECM component is laminin (LM). Laminin is found associated

with type IV collagen in the basal lamina. Each laminin molecule consists of three polypeptide

subunits (α−, β− and γ− chains) stabilized by disulfide bridges. Like FN, laminin can bind to

both ECM components and cell surface receptors and it is believed to influence cell attachment,

migration and differentiation104.

2.4.4 Synthetic Scaffold and Materials

A wide range of synthetic polymeric materials can be fabricated into scaffolds suitable

for cardiac tissue engineering. These scaffolds can be porous or fibrous in nature. By altering

the polymeric structure, the mechanical properties and biological responses of scaffolds can be

Page 35: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

23

fine tuned. Polyglycolic acid (PGA) and its copolymers with polylactic acid (PLA) and

polycaprolactone (PCL) form biodegradable scaffolds that have been used extensively in tissue

engineering applications. Rat neonatal CM seeded on PGA scaffold remain viable and

contractile when cultured in a spinner flask in vitro110. To better control the orientation of cell,

alignment polymer can be electrospun such that fibers are directionally arranged111.

Furthermore, bioactive molecules that enhance cell attachment112 and biodegradability113 can be

incorporated into the polymer structures during synthesis. Therefore, the resulting material can

inherit the mechanical advantages of synthetic polymers while gaining a more natural-like

cellular interface that is more suited for tissue development.

2.5 Fabrication and Culture Platforms for Cardiac Tissue Engineering

The interaction between cells and biomaterials on a molecular level is one of many

factors that will contribute to the functionality of the engineered tissue. To maximize the

regenerative potential of the cells, one must consider the impact of tissue fabrication and culture

strategies on the cells’ behavior.

Most biomaterial-cell combinations are fabricated by either seeding cells onto a pre-

fabricated substrate, or by mixing cells with the biomaterial prior to scaffold casting. Both

methods are currently used in the fabrication of cardiac tissue, and each has its benefits. The use

of pre-formed scaffolds allows the fabrication process to be uncoupled from the cell seeding and

permits the usage of processes that are not compatible with cells. Examples of these processes

include salt leaching, free radical polymerization, and plasma surface modifications. The wide

array of methods available for cell-free scaffold fabrication means that scaffold can have a wider

range of biological and mechanical properties. The disadvantage of this method is that cells can

only be seeded on the surface and must actively migrate into the core of the scaffolds. This may

lead to uneven cell seeding throughout the depth of the scaffold.

One way to improve the uniformity of seeding cells within the scaffold is to combine

cells with compatible hydrogel prior to casting and gelation. The Zimmermann group has

demonstrated that CM embedded in a collagen and Matrigel based hydrogel can be casted into

sheet114, ring115 or pouch116 forms; each with their unique structures and applications.

Nonetheless, cells embedded near the core of the gel still suffer from diffusion limitation and

tend to have low viability. Therefore, recent studies have shifted their focus towards the

Page 36: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

24

fabrications of microtissue such as the modular construct approach described within this thesis.

By shrinking the size of the scaffold down to several hundred micrometers in diameter, cells

embedded in the core may receive sufficient nutrients to survive. Kelm et al. have created

microtissues (~100 µm) containing CM in a hang drop culture system that are capable of

supporting core tissue viability117,118.

A variation on the modular scaffold design is cell sheet assembly. Many cells, including

fibroblast and CM, secret their own ECM components and form monolayer sheets when

cultured in vitro. Using a thermoresponsive poly N-Isopropylacrylamide coating to control the

growth of cell sheets, Shimizu et al. were able to cultivate contractile CM sheets and assemble

them into multilayer tissues119. The same group has also stacked sheets of fibroblast and EC

with CM sheets in alternating layers to improve angiogenesis when implanted in a rat model120.

Schematic diagrams of these cardiac tissue engineering strategies can be found in Figure 2-3.

Achieving adequate tissue perfusion and proper ECM architecture are essential for a

culture platform, but they are not enough to ensure the maintenance of phenotype and

functionality of cells 121. It is well known that primary cells can be successfully cultured and

expanded in vitro, but over time they lose cell-specific metabolic functions and cell-cell

interactions, this limiting their ability to organize into native-like, functional microstructures. In

native cardiac muscle, CM and CF are subjected to high level of mechanical stress during each

contraction cycle. Moreover, they are constantly generating and transmitting electrical impulses

over the myocardium. It is known that exposure to subtle mechanical stresses such as tensional

and shear on engineered cardiac tissues induces cell alignment, myosin fiber expression and

microstructure organization in vitro resembling native tissue 122. Electrical field stimulation

elicits a similar response in CM, and the expression of Cx43 corresponds to the direction of cell

alignment. Mechanical and electrical conditioning also results in a stronger contractile force,

although the magnitude is still much smaller than compared to native tissue123,124. These

evidences have illustrated that the delivery of physiologically relevant stimulus is crucial for

cardiac tissue development.

Page 37: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

25

Figure 2-3: Schematic diagram of the three major cardiac tissue engineering strategies. Cardiac

construct could be fabricated by seeding functional cells, such as CM and EC, onto pre-

fabricated scaffold. Alternatively, cells could be embedded into hydrogel and molded into the

desired geometry. An emerging technique for fabricating 3-D tissue is to stack 2-D cell sheets

together into a scaffold free constructs.

2.6 Angiogenesis and Cardiac Tissue Vascularization Strategies

The capacity to delivery sufficient nutrients to cells is one of the important determinants

to the viability and functionality of implanted tissue. The challenge in solving the mass transport

problem is obvious, considering that the first generation of successfully commercialized

artificial tissue comprise mainly of thin structures, such as skin grafts and corneas (in the

Page 38: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

26

millimeter thickness range), or avascular tissues like cartilage and tendons. There have been

continual clinical demand and interest in fabricating large, 3-dimensional tissues containing

metabolically functional cells. In particular, studies of angiogenesis and vasculogenesis in the

fields of tumor biology and developmental biology have been instrumental in the development

of novel strategies in scaffold vascularization.

Angiogenesis is the sprouting and growth of existing blood vessels into areas of poorly

vascularized tissues. This phenomenon has gained significant interest after tumor researchers

observed the in-growth of new vasculature into developing tumors. The process of angiogenesis

is well characterized and can be described in the following steps125. First, the parent blood

vessel undergoes vasodilation in order to disrupt interendothelial cell junctions. The sub-

endothelium basement membrane is then digested by secreted matrix metalloproteinases

(MMPs), followed by EC mobilization and the formation of a migration front of the new

capillary. The luminal space is formed by the morphological change of EC into tubules like

structure. Maturation of the new vessel is achieved through basement membrane synthesis and

recruitment of pericytes. In large caliber vessels, this pericyte layer is eventually replaced by

smooth muscle cells. On the other hand, the process of vasculogenesis does not require

sprouting from existence blood vessels. Instead, endothelial progenitor cells (EPC) known as

angioblasts begin to aggregate and establish cell-cell contact. The cell mass then gives rise to

nascent endothelial tubes that mature through the recruitment of pericytes. Although

vasculogenesis is the major mechanism of embryonic blood vessel formation, it has

demonstrated that EPC exist in the adult blood stream and may be involved in postnatal blood

vessel formation.

In both angiogenesis and vasculogenesis, the initiating events are driven by the presence

of angiogenic factors, usually in the form of growth factor such as vascular endothelial growth

factor (VEGF), basic fibroblast growth factor (b-FGF), transforming growth factor beta (TGF-β)

and platelet-derived growth factor (PDGF). Chemical species such as nitric oxide (NO) may

also serve as angiogenic factors. Some of the growth factors act directly on EC motility and

proliferation (VEGF and b-FGF). Others act indirectly (TGF-β and PDGF) on supporting cell

types that are involved in vessel maturation. In either case, the spatial and temporal distributions

of angiogenic factors are crucial to the growth rate, degree of branching and quality of new

blood vessels formed. Angiogenic factors are known to function synergistically via receptor

tyrosine kinase mediated downstream signaling pathways crosstalk126.

Page 39: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

27

Figure 2-4: Changes in the endothelium during angiogenesis. Cell-cell integration is maintained

by adherens junction proteins (VE-cadherin and PECAM), while the basal surfaces are anchored

to ECM composed of laminin and type IV collagen. In response to stimulus such as VEGF,

adherens junctions between EC are disassembled and EC begin to sprout toward the angiogenic

signals. At the same time, the underlying basement membrane is dissolved by proteolytic

enzymes and a provisional ECM is secreted by underlying cells to facilitate EC migration.

(Adapted from Stupack et al.127, 2004)

Early 3D cardiac scaffold designs relied on hypoxic environment and growth factor

secreted by embedded cells as angiogenic signals114,128,129. The rationale for this approach was

based on what was observed during tumor angiogenesis, where hypoxia induced growth factors

secretions were able to induce sufficient new blood vessel growth to sustain the tumor.

Page 40: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

28

However, this method is largely incompatible in tissue engineering application, since the time

frame of angiogenesis is usually measured in weeks, whereas most cells would not survive for

more than a day or two without sufficient perfusion.

One way to enhance the rate of vascularization is to incorporate EC into cardiac scaffolds

with the view that they can help initiate vasculogenesis and secrete growth factor to drive

angiogenesis. Variations of EC seeding strategies include static surface seeding and

homogenous EC embedding into 3D scaffolds. Seeding of EC on scaffolds is often combined

with some form of growth factors delivery (Ang-1, VEGF, b-FGF, TGF-β and PDGF) that are

designed to locally affect the angiogenic kinetics of implanted EC130. Controlled release of

VEGF has been shown to induce EC tubule formation131. A variation to embedding bioactive

molecules is to include naked plasmid DNA encoding VEGF or b-FGF in the scaffold. The idea

is to have seeded cells incorporate the naked plasmid and express the gene of interest thus

delivering the desired angiogenic effect132. However, this method is not widely used due to the

variability of VEGF expression. Besides DNA and growth factor embedded scaffold, transgenic

Chinese hamster ovary (CHO) cells expressing exogenous VEGF or b-FGF have been

encapsulated and co-implanted with cell seeded scaffold126. An obvious advantage of the

cellular protein expression method over bolus injection and controlled polymer release of

growth factor is the longevity and consistency of the growth factors delivered. This is especially

critical considering most growth factors and biological signaling molecules have a short half

life.

The rate of scaffold vascularization depends not only on growth factor and cells, but also

the material and design of the scaffold. Scaffolds made out of natural biomaterials can be easily

remodeled by cell and can also promote cell mobility through integrin binding. Therefore, the

ability of cells to remodel their surrounding may have an effect on their survival by influencing

the speed at which vascularization occurs.

2.7 In vivo models for evaluating engineered tissue

While in vitro assays provide a good starting point for the development of engineered

tissue, it is by no mean a good predictor of their in vivo behavior and performance.

Discrepancies between in vitro and in vivo observations are primarily due to the complex

interaction found in whole animals, including host immune responses, mechanical forces and

Page 41: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

29

extracellular matrices remodeling. Aside from species, animal models can be classified

according to their level of immune response toward the implanted tissue. A summary of recent

in vivo studies of engineered cardiac constructs is presented in Table 2-2. The choices of animal

models in these studies reflect the research question being addressed. Their features will be

discussed in the following section.

2.7.1 Allogeneic and immunosuppressed animal model

An allograft is an implant derived from a different individual of the same species. This is

by far the most common type of transplantation in human patients. The immune response

involved in allograft rejection is mediated by histocompatibility antigen on the donor cells, and

the most significant ones are the class I and II major histocompatibility complex (MHC). Class I

MHC is present on all cell types, while class II MHC is expressed on antigen presenting cells

(APC), namely dendritic cells, B cells and macrophages. Variations of MHC antigen between

individuals allow the host’s immune system to recognize self from non-self tissue. In general,

the severity of an allograft immune response is proportional to the divergence between the MHC

antigens.

An immune response can be triggered by MHC antigens via the binding of T cell

receptors and the subsequent activation of T cells. The process of T cell activation is further

regulated by co-stimulatory signaling molecules expressed on APCs. Once activated, T cells can

take on either CD4+ or CD8+ phenotype. T cells expressing CD4 molecule, also known as

helper T cells, function as a mediator of the immune response by promoting the proliferation

and cytotoxicity of macrophages and other T cells. The other type of T cells expressed CD8

molecule are known as cytotoxic T cell, and they are capable of causing cell death by releasing

cytokines or via direct Fas ligand binding with Fas receptors on target cells.

Even though allograft rejection is most prominent in whole organ transplants with intact

vasculature, the likelihood of initiating an immune response in engineered tissue derived from

allogeneic cells is significant enough to raise concerns when trying to achieve a controlled in

vivo model for studying tissue remodeling. In recent years, the development of genetically or

pharmacologically induced immunecompromised animal models allows researchers to

circumvent some of the challenges in host-versus-graft reactions. Genetically altered animals

such as athymic nude rats and severe combined immuno-deficient (SCID) mice lack the specific

Page 42: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

30

organ and cell types to effectively mount an adaptive immune response toward non-self tissues.

However, their disadvantage is that their immune system mutation may affect other organ

system and lead to other undesirable phenotypes, such as their increased susceptibility toward

tumor formation and shorter life span.

Another method of creating immunecompromised animal is by using pharmacological

agents to inhibit the activation and proliferation of host T cells and B cells. Immunosuppressants

such as azathioprine and mycophenolate mofetil act as antimetabolites to T and B cells. Others

such as cyclosporine and tacrolimus exert their action by inhibiting calcineurin mediated IL-2

expression by T cells. While long-term side effects commonly seen in human patients are

unlikely to be present during the typical implantation period of 1 to 4 weeks in animal models,

they should be taken into consideration and closely monitored to ensure that the observations are

not affected. Animals that are receiving immunosuppressants generally do not suffer from

aforementioned systemic problems that plague other immunocompromised animals. The uses of

pharmacological agents also allow researchers to titrate the desire amount of immune response

in the final model. This may be a useful way to study the effects that different immune

components have on the outcome of the implants. Finally, the use of immunosuppressant in

allotransplantation represents a model that closely resembles current clinical practice. The

results generated from these types of studies can be more easily translated into bedside

applications.

Page 43: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

31

Cel

l typ

e Im

plan

t typ

e A

nim

al M

odel

E

ndpo

int

Key

resu

lts

Neo

nata

l rat

CM

Pe

ri-in

farc

t inj

ectio

n

Rat

- sy

ngen

eic

4 w

eeks

Im

plan

ted

CM

exp

ress

CX

-43

(200

4)4

Neo

nata

l rat

CM

C

olla

gen

+ M

atrig

el ri

ng-

type

EH

T R

at -

syng

enei

c 2

wee

ks

Imm

unos

uppr

esse

nt n

eces

sary

for E

HT

surv

ival

in v

ivo.

EH

T he

avily

va

scua

lrize

d an

d tis

sue

mat

urat

ion

obse

rved

in v

ivo

(200

2)13

3 . N

eona

tal r

at C

M

Alg

inat

e sc

affo

ld

Rat

– a

llogr

aft,

4 w

eeks

Pr

evas

cual

rized

scaf

fold

s stru

ctur

ally

and

ele

ctric

ally

inte

grat

ed in

to

host

myo

card

ium

. Als

o th

icke

n sc

ar ti

ssue

and

pre

vent

s fur

ther

LV

di

latio

n (2

009)

134

Skel

etal

myo

blas

t C

olla

gen

+ M

atrig

el p

atch

ov

er M

I R

at -

syng

enei

c 4

wee

ks

Stro

ng im

mun

e re

spon

se. I

mpr

oved

syst

olic

func

tion,

but

no

indi

catio

n of

impl

ante

d ce

ll su

rviv

al (2

008)

135

hESC

-CM

, em

bryo

nic

FB a

nd

HU

VEC

PLLA

/PLG

A sc

affo

ld

Rat

– a

llogr

aft,

imm

unos

uppr

esse

d

2 w

eeks

So

me

mat

urat

ion

of C

M in

to st

riate

d st

ruct

ure

and

expr

esse

s CX

-43.

Fo

rmat

ion

of d

onor

and

hos

t der

ived

blo

od v

esse

ls (2

010)

136

hESC

der

ived

CM

, FB

and

EC

M

atrix

-fre

e sh

eets

ove

r he

alth

y he

art

Rat

– a

llogr

aft,

athy

mic

hos

t 1

wee

k Fo

rmat

ion

of b

lood

ves

sel-l

ike

stru

ctur

es. M

HC

and

Nkx

-2.5

pos

itive

C

M fo

und

in p

atch

(200

9)13

7 N

eona

tal r

at C

M

Col

lage

n m

esh

into

skel

etal

m

uscl

e po

uch

Rat

– a

llogr

aft,

imm

unos

uppr

esse

d

2,3

and4

w

eeks

C

M su

rviv

ed a

t 4 w

eeks

, but

did

not

form

stria

ted

bund

les.

Hos

t ve

ssel

aro

und

perip

hery

at 4

wee

ks (2

008)

138 .

Neo

nata

l rat

CM

and

H

UV

EC

ECM

-fre

e m

icro

tissu

e in

ject

ed in

to p

eric

ardi

al

cavi

ty

Rat

- al

logr

aft

1 w

eek

Mic

rotis

sue

inte

grat

ed in

to h

ost m

yoca

rdiu

m a

s wel

l as t

issu

e el

onga

tion

by d

ay 7

(200

6)13

9

Neo

nata

l rat

CM

C

olla

gen

+ M

atrig

el b

and-

like

EHT

over

MI

Rat

– a

llogr

aft,

imm

unos

uppr

esse

d

4 w

eeks

Fo

rmat

ion

of h

ighl

y di

ffer

entia

ted

card

iac

mus

cle

over

MI s

car.

Ves

sel s

truct

ures

foun

d w

ithin

EH

T. Im

prov

e sy

stol

ic a

nd d

iast

olic

fu

nctio

n (2

006)

140

Neo

nata

l rat

CM

C

olla

gen

+ M

atrig

el p

ouch

-lik

e EH

T ov

er h

ealth

y he

art

Rat

– a

llogr

aft,

imm

unos

uppr

esse

d

2 w

eeks

A

ggre

gatio

n of

CM

nex

t to

host

myo

card

ium

but

sepa

rate

d by

cel

l-fr

ee g

ap. C

M fo

rm lo

ose

by d

iffer

entia

ted

mus

cle.

Abu

ndan

t CX

43

expr

essi

on (2

007)

116 .

Neo

nata

l rat

CM

C

olla

gen

+ M

atrig

el

inje

ctio

n R

at –

allo

graf

t, im

mun

osup

pres

sed

4 w

eeks

C

M su

rviv

ed a

nd e

xpre

sses

CX

43, b

ut n

o st

riate

d tis

sue

form

atio

n (2

006)

141 .

Rat

CM

and

EC

m

atrix

-fre

e sh

eets

R

at –

allo

graf

t, at

hym

ic h

ost

4 w

eeks

Im

prov

ed v

esse

l den

sity

in c

o-cu

lture

pat

ch14

2 Im

plan

ted

EC in

filtra

ted

into

hos

t myo

card

ium

and

form

ves

sels

(2

008)

142

Tab

le 2

-2: A

sum

mar

y of

rece

nt im

plan

tatio

n st

udie

s usi

ng v

ario

us c

ardi

ac ti

ssue

s eng

inee

ring

stra

tegi

es.

Page 44: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

32

Reference List

1. L. Bu, X. Jiang, S. Martin-Puig, L. Caron, S. Zhu, Y. Shao, D. J. Roberts, P. L. Huang, I.

J. Domian, and K. R. Chien, Human ISL1 heart progenitors generate diverse multipotent

cardiovascular cell lineages. Nature 460, 113-117 (2009).

2. H. Kubo, N. Jaleel, A. Kumarapeli, R. M. Berretta, G. Bratinov, X. Shan, H. Wang, S. R.

Houser, and K. B. Margulies, Increased cardiac myocyte progenitors in failing human

hearts. Circulation 118, 649-657 (2008).

3. L. C. Amado, A. P. Saliaris, K. H. Schuleri, M. St John, J. S. Xie, S. Cattaneo, D. J.

Durand, T. Fitton, J. Q. Kuang, G. Stewart, S. Lehrke, W. W. Baumgartner, B. J. Martin,

A. W. Heldman, and J. M. Hare, Cardiac repair with intramyocardial injection of

allogeneic mesenchymal stem cells after myocardial infarction. Proc. Natl. Acad. Sci. U.

S. A 102, 11474-11479 (2005).

4. M. Yoshiyama, T. Hayashi, Y. Nakamura, T. Omura, Y. Izumi, R. Matsumoto, K.

Takeuchi, Y. Kitaura, and J. Yoshikawa, Effects of cellular cardiomyoplasty on

ventricular remodeling assessed by Doppler echocardiography and topographic

immunohistochemistry. Circ. J. 68, 580-586 (2004).

5. P. Farahmand, T. Y. Lai, R. D. Weisel, S. Fazel, T. Yau, P. Menasche, and R. K. Li,

Skeletal myoblasts preserve remote matrix architecture and global function when

implanted early or late after coronary ligation into infarcted or remote myocardium.

Circulation 118, S130-S137 (2008).

6. T. Patila, T. Ikonen, E. Kankuri, A. Uutela, J. Lommi, L. Krogerus, P. Salmenpera, J.

Bizik, K. Lauerma, and A. Harjula, Improved diastolic function after myoblast

transplantation in a model of ischemia-infarction. Scand. Cardiovasc. J. 43, 100-109

(2009).

Page 45: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

33

7. W. Sherman, K. L. He, G. H. Yi, J. Wang, J. Harvey, M. J. Lee, H. Haimes, P. Lee, E.

Miranda, S. Kanwal, and D. Burkhoff, Myoblast transfer in ischemic heart failure:

effects on rhythm stability. Cell Transplant. 18, 333-341 (2009).

8. M. A. Laflamme, K. Y. Chen, A. V. Naumova, V. Muskheli, J. A. Fugate, S. K. Dupras,

H. Reinecke, C. Xu, M. Hassanipour, S. Police, C. O'Sullivan, L. Collins, Y. Chen, E.

Minami, E. A. Gill, S. Ueno, C. Yuan, J. Gold, and C. E. Murry, Cardiomyocytes

derived from human embryonic stem cells in pro-survival factors enhance function of

infarcted rat hearts. Nat. Biotechnol. 25, 1015-1024 (2007).

9. K. R. Stevens, K. L. Kreutziger, S. K. Dupras, F. S. Korte, M. Regnier, V. Muskheli, M.

B. Nourse, K. Bendixen, H. Reinecke, and C. E. Murry, Physiological function and

transplantation of scaffold-free and vascularized human cardiac muscle tissue. Proc.

Natl. Acad. Sci. U. S. A 106, 16568-16573 (2009).

10. J. A. Mund, D. A. Ingram, M. C. Yoder, and J. Case, Endothelial progenitor cells and

cardiovascular cell-based therapies. Cytotherapy. 11, 103-113 (2009).

11. M. A. Laflamme, S. Zbinden, S. E. Epstein, and C. E. Murry, Cell-based therapy for

myocardial ischemia and infarction: pathophysiological mechanisms. Annu. Rev. Pathol.

2, 307-339 (2007).

12. L. A. Benvenuti, L. F. Moreira, V. D. Aiello, and M. L. Higuchi, Sequential histologic

analysis of the myocardium after dynamic cardiomyoplasty: a study based on right

ventricular endomyocardial biopsies. J. Heart Lung Transplant. 23, 1438-1440 (2004).

13. D. Schmidt, A. Mol, S. Neuenschwander, C. Breymann, M. Gossi, G. Zund, M. Turina,

and S. P. Hoerstrup, Living patches engineered from human umbilical cord derived

fibroblasts and endothelial progenitor cells. Eur. J. Cardiothorac. Surg. 27, 795-800

(2005).

14. W. Risau, Differentiation of endothelium. FASEB J. 9, 926-933 (1995).

Page 46: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

34

15. C. Garlanda and E. Dejana, Heterogeneity of endothelial cells. Specific markers.

Arterioscler. Thromb. Vasc. Biol. 17, 1193-1202 (1997).

16. C. E. Murry and G. Keller, Differentiation of embryonic stem cells to clinically relevant

populations: lessons from embryonic development. Cell 132, 661-680 (2008).

17. K. R. Chien, I. J. Domian, and K. K. Parker, Cardiogenesis and the complex biology of

regenerative cardiovascular medicine. Science 322, 1494-1497 (2008).

18. J. Nussbaum, E. Minami, M. A. Laflamme, J. A. Virag, C. B. Ware, A. Masino, V.

Muskheli, L. Pabon, H. Reinecke, and C. E. Murry, Transplantation of undifferentiated

murine embryonic stem cells in the heart: teratoma formation and immune response.

FASEB J. 21, 1345-1357 (2007).

19. V. C. Chen, R. Stull, D. Joo, X. Cheng, and G. Keller, Notch signaling respecifies the

hemangioblast to a cardiac fate. Nat. Biotechnol. 26, 1169-1178 (2008).

20. J. Ellis, B. G. Bruneau, G. Keller, I. R. Lemischka, A. Nagy, J. Rossant, D. Srivastava,

P. W. Zandstra, and W. L. Stanford, Alternative induced pluripotent stem cell

characterization criteria for in vitro applications. Cell Stem Cell 4, 198-199 (2009).

21. D. C. Kirouac and P. W. Zandstra, The systematic production of cells for cell therapies.

Cell Stem Cell 3, 369-381 (2008).

22. M. A. Laflamme, J. Gold, C. Xu, M. Hassanipour, E. Rosler, S. Police, V. Muskheli, and

C. E. Murry, Formation of human myocardium in the rat heart from human embryonic

stem cells. Am. J. Pathol. 167, 663-671 (2005).

23. K. R. Stevens, L. Pabon, V. Muskheli, and C. E. Murry, Scaffold-free human cardiac

tissue patch created from embryonic stem cells. Tissue Eng Part A 15, 1211-1222

(2009).

24. K. Takahashi and S. Yamanaka, Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676 (2006).

Page 47: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

35

25. G. Vunjak-Novakovic, N. Tandon, A. Godier, R. Maidhof, A. Marsano, T. P. Martens,

and M. Radisic, Challenges in cardiac tissue engineering. Tissue Eng Part B Rev. 16,

169-187 (2010).

26. R. Madonna and R. De Caterina, Adipose tissue: a new source for cardiovascular repair.

J. Cardiovasc. Med. (Hagerstown. ) 11, 71-80 (2010).

27. A. J. Sloan and R. J. Waddington, Dental pulp stem cells: what, where, how? Int. J.

Paediatr. Dent. 19, 61-70 (2009).

28. X. Guan, D. M. Delo, A. Atala, and S. Soker, In vitro cardiomyogenic potential of

human amniotic fluid stem cells. J. Tissue Eng Regen. Med. (2010).

29. E. Galende, I. Karakikes, L. Edelmann, R. J. Desnick, T. Kerenyi, G. Khoueiry, J.

Lafferty, J. T. McGinn, M. Brodman, V. Fuster, R. J. Hajjar, and K. Polgar, Amniotic

fluid cells are more efficiently reprogrammed to pluripotency than adult cells. Cell

Reprogram. 12, 117-125 (2010).

30. S. J. Gwak, S. H. Bhang, H. S. Yang, S. S. Kim, D. H. Lee, S. H. Lee, and B. S. Kim, In

vitro cardiomyogenic differentiation of adipose-derived stromal cells using transforming

growth factor-beta1. Cell Biochem. Funct. 27, 148-154 (2009).

31. L. Zhang and C. Chan, Isolation and enrichment of rat mesenchymal stem cells (MSCs)

and separation of single-colony derived MSCs. J. Vis. Exp. (2010).

32. S. H. Bhang, S. J. Gwak, T. J. Lee, S. S. Kim, H. H. Park, M. H. Park, D. H. Lee, S. H.

Lee, and B. S. Kim, Cyclic mechanical strain promotes transforming-growth-factor-

beta1-mediated cardiomyogenic marker expression in bone-marrow-derived

mesenchymal stem cells in vitro. Biotechnol. Appl. Biochem. 55, 191-197 (2010).

33. A. Cselenyak, E. Pankotai, E. M. Horvath, L. Kiss, and Z. Lacza, Mesenchymal stem

cells rescue cardiomyoblasts from cell death in an in vitro ischemia model via direct cell-

to-cell connections. BMC. Cell Biol. 11, 29 (2010).

Page 48: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

36

34. V. Schachinger, S. Erbs, A. Elsasser, W. Haberbosch, R. Hambrecht, H. Holschermann,

J. Yu, R. Corti, D. G. Mathey, C. W. Hamm, T. Suselbeck, B. Assmus, T. Tonn, S.

Dimmeler, and A. M. Zeiher, Intracoronary bone marrow-derived progenitor cells in

acute myocardial infarction. N. Engl. J. Med. 355, 1210-1221 (2006).

35. S. R. Li, X. Y. Qi, F. L. Hu, J. Q. Zhang, T. H. Wang, Y. Dang, C. L. Meng, H. L. Liu,

Y. X. Li, D. Wu, J. Dong, L. Y. Xun, L. H. Gao, and F. C. Jin, Mechanisms of

improvement of left ventricle remodeling by trans-planting two kinds of autologous bone

marrow stem cells in pigs. Chin Med. J. (Engl. ) 121, 2403-2409 (2008).

36. C. H. Yoon, M. Koyanagi, K. Iekushi, F. Seeger, C. Urbich, A. M. Zeiher, and S.

Dimmeler, Mechanism of Improved Cardiac Function After Bone Marrow Mononuclear

Cell Therapy. Role of Cardiovascular Lineage Commitment. Circulation (2010).

37. S. Zhang, P. Zhang, J. Guo, Z. Jia, K. Ma, Y. Liu, C. Zhou, and L. Li, Enhanced

cytoprotection and angiogenesis by bone marrow cell transplantation may contribute to

improved ischemic myocardial function. Eur. J. Cardiothorac. Surg. 25, 188-195

(2004).

38. M. A. Laflamme, S. Zbinden, S. E. Epstein, and C. E. Murry, Cell-based therapy for

myocardial ischemia and infarction: pathophysiological mechanisms. Annu. Rev. Pathol.

2, 307-339 (2007).

39. P. A. Zuk, M. Zhu, P. Ashjian, D. A. De Ugarte, J. I. Huang, H. Mizuno, Z. C. Alfonso,

J. K. Fraser, P. Benhaim, and M. H. Hedrick, Human adipose tissue is a source of

multipotent stem cells. Mol. Biol. Cell 13, 4279-4295 (2002).

40. Y. H. Song, S. Gehmert, S. Sadat, K. Pinkernell, X. Bai, N. Matthias, and E. Alt, VEGF

is critical for spontaneous differentiation of stem cells into cardiomyocytes. Biochem.

Biophys. Res. Commun. 354, 999-1003 (2007).

41. K. G. Gaustad, A. C. Boquest, B. E. Anderson, A. M. Gerdes, and P. Collas,

Differentiation of human adipose tissue stem cells using extracts of rat cardiomyocytes.

Biochem. Biophys. Res. Commun. 314, 420-427 (2004).

Page 49: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

37

42. M. Peran, J. A. Marchal, E. Lopez, M. Jimenez-Navarro, H. Boulaiz, F. Rodriguez-

Serrano, E. Carrillo, G. Sanchez-Espin, E. de Teresa, D. Tosh, and A. Aranega, Human

cardiac tissue induces transdifferentiation of adult stem cells towards cardiomyocytes.

Cytotherapy. 12, 332-337 (2010).

43. M. Jumabay, T. Matsumoto, S. Yokoyama, K. Kano, Y. Kusumi, T. Masuko, M.

Mitsumata, S. Saito, A. Hirayama, H. Mugishima, and N. Fukuda, Dedifferentiated fat

cells convert to cardiomyocyte phenotype and repair infarcted cardiac tissue in rats. J.

Mol. Cell Cardiol. 47, 565-575 (2009).

44. K. Rubina, N. Kalinina, A. Efimenko, T. Lopatina, V. Melikhova, Z. Tsokolaeva, V.

Sysoeva, V. Tkachuk, and Y. Parfyonova, Adipose stromal cells stimulate angiogenesis

via promoting progenitor cell differentiation, secretion of angiogenic factors, and

enhancing vessel maturation. Tissue Eng Part A 15, 2039-2050 (2009).

45. X. Bai, Y. Yan, Y. H. Song, M. Seidensticker, B. Rabinovich, R. Metzele, J. A.

Bankson, D. Vykoukal, and E. Alt, Both cultured and freshly isolated adipose tissue-

derived stem cells enhance cardiac function after acute myocardial infarction. Eur. Heart

J. 31, 489-501 (2010).

46. S. Hwangbo, J. Kim, S. Her, H. Cho, and J. Lee, Therapeutic potential of human adipose

stem cells in a rat myocardial infarction model. Yonsei Med. J. 51, 69-76 (2010).

47. S. Seki, M. Nagashima, Y. Yamada, M. Tsutsuura, T. Kobayashi, A. Namiki, and N.

Tohse, Fetal and postnatal development of Ca2+ transients and Ca2+ sparks in rat

cardiomyocytes. Cardiovasc. Res. 58, 535-548 (2003).

48. J. B. Delcarpio, W. C. Claycomb, and R. L. Moses, Ultrastructural morphometric

analysis of cultured neonatal and adult rat ventricular cardiac muscle cells. Am. J. Anat.

186, 335-345 (1989).

49. B. E. Sumpio, J. T. Riley, and A. Dardik, Cells in focus: endothelial cell. Int. J. Biochem.

Cell Biol. 34, 1508-1512 (2002).

Page 50: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

38

50. E. Arciniegas, L. Ponce, Y. Hartt, A. Graterol, and R. G. Carlini, Intimal thickening

involves transdifferentiation of embryonic endothelial cells. Anat. Rec. 258, 47-57

(2000).

51. M. G. Frid, V. A. Kale, and K. R. Stenmark, Mature vascular endothelium can give rise

to smooth muscle cells via endothelial-mesenchymal transdifferentiation: in vitro

analysis. Circ. Res. 90, 1189-1196 (2002).

52. G. Paranya, S. Vineberg, E. Dvorin, S. Kaushal, S. J. Roth, E. Rabkin, F. J. Schoen, and

J. Bischoff, Aortic valve endothelial cells undergo transforming growth factor-beta-

mediated and non-transforming growth factor-beta-mediated transdifferentiation in vitro.

Am. J. Pathol. 159, 1335-1343 (2001).

53. W. C. Aird, Endothelium as an organ system. Crit Care Med. 32, S271-S279 (2004).

54. G. J. Broze, Jr., Tissue factor pathway inhibitor. Thromb. Haemost. 74, 90-93 (1995).

55. J. B. Hansen, B. Svensson, R. Olsen, M. Ezban, B. Osterud, and R. H. Paulssen, Heparin

induces synthesis and secretion of tissue factor pathway inhibitor from endothelial cells

in vitro. Thromb. Haemost. 83, 937-943 (2000).

56. A. D. Westmuckett, C. Lupu, S. Roquefeuil, T. Krausz, V. V. Kakkar, and F. Lupu,

Fluid flow induces upregulation of synthesis and release of tissue factor pathway

inhibitor in vitro. Arterioscler. Thromb. Vasc. Biol. 20, 2474-2482 (2000).

57. C. Lupu, F. Lupu, U. Dennehy, V. V. Kakkar, and M. F. Scully, Thrombin induces the

redistribution and acute release of tissue factor pathway inhibitor from specific granules

within human endothelial cells in culture. Arterioscler. Thromb. Vasc. Biol. 15, 2055-

2062 (1995).

58. T. Bombeli, M. Mueller, and A. Haeberli, Anticoagulant properties of the vascular

endothelium. Thromb. Haemost. 77, 408-423 (1997).

Page 51: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

39

59. C. T. Esmon, Thrombomodulin as a model of molecular mechanisms that modulate

protease specificity and function at the vessel surface. FASEB J. 9, 946-955 (1995).

60. C. T. Esmon, The endothelial cell protein C receptor. Thromb. Haemost. 83, 639-643

(2000).

61. T. A. Drake, J. H. Morrissey, and T. S. Edgington, Selective cellular expression of tissue

factor in human tissues. Implications for disorders of hemostasis and thrombosis. Am. J.

Pathol. 134, 1087-1097 (1989).

62. H. Holzmuller, T. Moll, R. Hofer-Warbinek, D. Mechtcheriakova, B. R. Binder, and E.

Hofer, A transcriptional repressor of the tissue factor gene in endothelial cells.

Arterioscler. Thromb. Vasc. Biol. 19, 1804-1811 (1999).

63. G. M. Riha, P. H. Lin, A. B. Lumsden, Q. Yao, and C. Chen, Roles of hemodynamic

forces in vascular cell differentiation. Ann. Biomed. Eng 33, 772-779 (2005).

64. W. C. Aird, Endothelial cell heterogeneity. Crit Care Med. 31, S221-S230 (2003).

65. M. Papadaki, S. G. Eskin, J. Ruef, M. S. Runge, and L. V. McIntire, Fluid shear stress as

a regulator of gene expression in vascular cells: possible correlations with diabetic

abnormalities. Diabetes Res. Clin. Pract. 45, 89-99 (1999).

66. N. J. Boudreau and P. L. Jones, Extracellular matrix and integrin signalling: the shape of

things to come. Biochem. J. 339 ( Pt 3), 481-488 (1999).

67. E. Tzima, M. Irani-Tehrani, W. B. Kiosses, E. Dejana, D. A. Schultz, B. Engelhardt, G.

Cao, H. DeLisser, and M. A. Schwartz, A mechanosensory complex that mediates the

endothelial cell response to fluid shear stress. Nature 437, 426-431 (2005).

68. H. J. Schnittler, Structural and functional aspects of intercellular junctions in vascular

endothelium. Basic Res. Cardiol. 93 Suppl 3, 30-39 (1998).

Page 52: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

40

69. Y. J. Chiu, K. Kusano, T. N. Thomas, and K. Fujiwara, Endothelial cell-cell adhesion

and mechanosignal transduction. Endothelium 11, 59-73 (2004).

70. L. Gonzalez-Mariscal, A. Betanzos, P. Nava, and B. E. Jaramillo, Tight junction

proteins. Prog. Biophys. Mol. Biol. 81, 1-44 (2003).

71. A. S. Yap, W. M. Brieher, and B. M. Gumbiner, Molecular and functional analysis of

cadherin-based adherens junctions. Annu. Rev. Cell Dev. Biol. 13, 119-146 (1997).

72. M. G. Lampugnani, M. Corada, P. Andriopoulou, S. Esser, W. Risau, and E. Dejana,

Cell confluence regulates tyrosine phosphorylation of adherens junction components in

endothelial cells. J. Cell Sci. 110 ( Pt 17), 2065-2077 (1997).

73. R. K. Jain, Molecular regulation of vessel maturation. Nat. Med. 9, 685-693 (2003).

74. S. Esser, M. G. Lampugnani, M. Corada, E. Dejana, and W. Risau, Vascular endothelial

growth factor induces VE-cadherin tyrosine phosphorylation in endothelial cells. J. Cell

Sci. 111 ( Pt 13), 1853-1865 (1998).

75. P. Carmeliet, M. G. Lampugnani, L. Moons, F. Breviario, V. Compernolle, F. Bono, G.

Balconi, R. Spagnuolo, B. Oostuyse, M. Dewerchin, A. Zanetti, A. Angellilo, V. Mattot,

D. Nuyens, E. Lutgens, F. Clotman, M. C. de Ruiter, A. Gittenberger-de Groot, R.

Poelmann, F. Lupu, J. M. Herbert, D. Collen, and E. Dejana, Targeted deficiency or

cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated

endothelial survival and angiogenesis. Cell 98, 147-157 (1999).

76. C. A. Souders, S. L. Bowers, and T. A. Baudino, Cardiac fibroblast: the renaissance cell.

Circ. Res. 105, 1164-1176 (2009).

77. S. W. van den Borne, J. Diez, W. M. Blankesteijn, J. Verjans, L. Hofstra, and J. Narula,

Myocardial remodeling after infarction: the role of myofibroblasts. Nat. Rev. Cardiol. 7,

30-37 (2010).

Page 53: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

41

78. P. Kohl, Heterogeneous cell coupling in the heart: an electrophysiological role for

fibroblasts. Circ. Res. 93, 381-383 (2003).

79. C. Louault, N. Benamer, J. F. Faivre, D. Potreau, and J. Bescond, Implication of

connexins 40 and 43 in functional coupling between mouse cardiac fibroblasts in

primary culture. Biochim. Biophys. Acta 1778, 2097-2104 (2008).

80. J. J. Tomasek, G. Gabbiani, B. Hinz, C. Chaponnier, and R. A. Brown, Myofibroblasts

and mechano-regulation of connective tissue remodelling. Nat. Rev. Mol. Cell Biol. 3,

349-363 (2002).

81. R. Kakkar and R. T. Lee, Intramyocardial fibroblast myocyte communication. Circ. Res.

106, 47-57 (2010).

82. D. Baronas-Lowell, J. L. Lauer-Fields, and G. B. Fields, Induction of endothelial cell

activation by a triple helical alpha2beta integrin ligand, derived from type I collagen

alpha1(I)496-507. J. Biol. Chem. 279, 952-962 (2004).

83. C. Garlanda and E. Dejana, Heterogeneity of endothelial cells. Specific markers.

Arterioscler. Thromb. Vasc. Biol. 17, 1193-1202 (1997).

84. E. W. Raines, The extracellular matrix can regulate vascular cell migration, proliferation,

and survival: relationships to vascular disease. Int. J. Exp. Pathol. 81, 173-182 (2000).

85. T. Ichii, H. Koyama, S. Tanaka, S. Kim, A. Shioi, Y. Okuno, E. W. Raines, H. Iwao, S.

Otani, and Y. Nishizawa, Fibrillar collagen specifically regulates human vascular

smooth muscle cell genes involved in cellular responses and the pericellular matrix

environment. Circ. Res. 88, 460-467 (2001).

86. H. Hutchings, N. Ortega, and J. Plouet, Extracellular matrix-bound vascular endothelial

growth factor promotes endothelial cell adhesion, migration, and survival through

integrin ligation. FASEB J. 17, 1520-1522 (2003).

Page 54: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

42

87. A. Orecchia, P. M. Lacal, C. Schietroma, V. Morea, G. Zambruno, and C. M. Failla,

Vascular endothelial growth factor receptor-1 is deposited in the extracellular matrix by

endothelial cells and is a ligand for the alpha 5 beta 1 integrin. J. Cell Sci. 116, 3479-

3489 (2003).

88. H. C. Ott, T. S. Matthiesen, S. K. Goh, L. D. Black, S. M. Kren, T. I. Netoff, and D. A.

Taylor, Perfusion-decellularized matrix: using nature's platform to engineer a

bioartificial heart. Nat. Med. 14, 213-221 (2008).

89. R. D. Harkness, Collagen. Sci. Prog. 54, 257-274 (1966).

90. D. J. Prockop and K. I. Kivirikko, Collagens: molecular biology, diseases, and potentials

for therapy. Annu. Rev. Biochem. 64, 403-434 (1995).

91. Carl Branden and John Tooze, Introduction to Protein Structure, (1998).

92. L. Espira and M. P. Czubryt, Emerging concepts in cardiac matrix biology. Can. J.

Physiol Pharmacol. 87, 996-1008 (2009).

93. C. H. Lee, A. Singla, and Y. Lee, Biomedical applications of collagen. Int. J. Pharm.

221, 1-22 (2001).

94. N. J. Vardaxis, M. E. Boon, and J. M. Ruijgrok, Calcification of cross-linked collagen-

elastin membrane implants in vivo and their proposed use in bone regeneration.

Biomaterials 17, 1489-1497 (1996).

95. A. H. Reddi, Morphogenesis and tissue engineering of bone and cartilage: inductive

signals, stem cells, and biomimetic biomaterials. Tissue Eng 6, 351-359 (2000).

96. M. Murata, B. Z. Huang, T. Shibata, S. Imai, N. Nagai, and M. Arisue, Bone

augmentation by recombinant human BMP-2 and collagen on adult rat parietal bone. Int.

J. Oral Maxillofac. Surg. 28, 232-237 (1999).

Page 55: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

43

97. M. L. Kahn, Platelet-collagen responses: molecular basis and therapeutic promise.

Semin. Thromb. Hemost. 30, 419-425 (2004).

98. W. A. Hsueh, R. E. Law, and Y. S. Do, Integrins, adhesion, and cardiac remodeling.

Hypertension 31, 176-180 (1998).

99. I. J. LeGrice, B. H. Smaill, L. Z. Chai, S. G. Edgar, J. B. Gavin, and P. J. Hunter,

Laminar structure of the heart: ventricular myocyte arrangement and connective tissue

architecture in the dog. Am. J. Physiol 269, H571-H582 (1995).

100. A. J. Pope, G. B. Sands, B. H. Smaill, and I. J. LeGrice, Three-dimensional transmural

organization of perimysial collagen in the heart. Am. J. Physiol Heart Circ. Physiol 295,

H1243-H1252 (2008).

101. P. M. Nielsen, I. J. Le Grice, B. H. Smaill, and P. J. Hunter, Mathematical model of

geometry and fibrous structure of the heart. Am. J. Physiol 260, H1365-H1378 (1991).

102. B. A. Coppola and J. H. Omens, Role of tissue structure on ventricular wall mechanics.

Mol. Cell Biomech. 5, 183-196 (2008).

103. A. Sosnik and M. V. Sefton, Semi-synthetic collagen/poloxamine matrices for tissue

engineering. Biomaterials 26, 7425-7435 (2005).

104. Bruce Alberts, Alexander Johnson, Julian Lewis, Martin Raff, Keith Roberts, and Peter

Walter, Cell Junctions, Cell Adhesion, and the Extracellular Matrix. In Molecular

Biology of Cell, 4th Edn, Garland Science, (2002).

105. A. Hinek and M. Rabinovitch, 67-kD elastin-binding protein is a protective "companion"

of extracellular insoluble elastin and intracellular tropoelastin. J. Cell Biol. 126, 563-574

(1994).

106. B. Li and V. Daggett, Molecular basis for the extensibility of elastin. J. Muscle Res. Cell

Motil. 23, 561-573 (2002).

Page 56: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

44

107. L. Debelle and A. M. Tamburro, Elastin: molecular description and function. Int. J.

Biochem. Cell Biol. 31, 261-272 (1999).

108. F. W. Keeley, C. M. Bellingham, and K. A. Woodhouse, Elastin as a self-organizing

biomaterial: use of recombinantly expressed human elastin polypeptides as a model for

investigations of structure and self-assembly of elastin. Philos. Trans. R. Soc. Lond B

Biol. Sci. 357, 185-189 (2002).

109. J. E. Schwarzbauer, Fibronectin: from gene to protein. Curr. Opin. Cell Biol. 3, 786-791

(1991).

110. R. L. Carrier, M. Papadaki, M. Rupnick, F. J. Schoen, N. Bursac, R. Langer, L. E. Freed,

and G. Vunjak-Novakovic, Cardiac tissue engineering: cell seeding, cultivation

parameters, and tissue construct characterization. Biotechnol. Bioeng. 64, 580-589

(1999).

111. M. Shin, O. Ishii, T. Sueda, and J. P. Vacanti, Contractile cardiac grafts using a novel

nanofibrous mesh. Biomaterials 25, 3717-3723 (2004).

112. T. G. Kim and T. G. Park, Biomimicking extracellular matrix: cell adhesive RGD

peptide modified electrospun poly(D,L-lactic-co-glycolic acid) nanofiber mesh. Tissue

Eng 12, 221-233 (2006).

113. I. C. Parrag and K. A. Woodhouse, Development of biodegradable polyurethane

scaffolds using amino acid and dipeptide-based chain extenders for soft tissue

engineering. J. Biomater. Sci. Polym. Ed 21, 843-862 (2010).

114. W. H. Zimmermann, C. Fink, D. Kralisch, U. Remmers, J. Weil, and T. Eschenhagen,

Three-dimensional engineered heart tissue from neonatal rat cardiac myocytes.

Biotechnol. Bioeng. 68, 106-114 (2000).

115. H. Naito, I. Melnychenko, M. Didie, K. Schneiderbanger, P. Schubert, S. Rosenkranz, T.

Eschenhagen, and W. H. Zimmermann, Optimizing engineered heart tissue for

therapeutic applications as surrogate heart muscle. Circulation 114, I72-I78 (2006).

Page 57: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

45

116. Y. Yildirim, H. Naito, M. Didie, B. C. Karikkineth, D. Biermann, T. Eschenhagen, and

W. H. Zimmermann, Development of a biological ventricular assist device: preliminary

data from a small animal model. Circulation 116, I16-I23 (2007).

117. J. M. Kelm, E. Ehler, L. K. Nielsen, S. Schlatter, J. C. Perriard, and M. Fussenegger,

Design of artificial myocardial microtissues. Tissue Eng 10, 201-214 (2004).

118. J. M. Kelm, V. Djonov, L. M. Ittner, D. Fluri, W. Born, S. P. Hoerstrup, and M.

Fussenegger, Design of custom-shaped vascularized tissues using microtissue spheroids

as minimal building units. Tissue Eng 12, 2151-2160 (2006).

119. T. Shimizu, M. Yamato, Y. Isoi, T. Akutsu, T. Setomaru, K. Abe, A. Kikuchi, M.

Umezu, and T. Okano, Fabrication of pulsatile cardiac tissue grafts using a novel 3-

dimensional cell sheet manipulation technique and temperature-responsive cell culture

surfaces. Circ. Res. 90, e40 (2002).

120. H. Kobayashi, T. Shimizu, M. Yamato, K. Tono, H. Masuda, T. Asahara, H. Kasanuki,

and T. Okano, Fibroblast sheets co-cultured with endothelial progenitor cells improve

cardiac function of infarcted hearts. J. Artif. Organs 11, 141-147 (2008).

121. Y. S. Zhao, C. Y. Wang, D. X. Li, X. Z. Zhang, Y. Qiao, X. M. Guo, X. L. Wang, C. M.

Dun, L. Z. Dong, and Y. Song, Construction of a unidirectionally beating 3-dimensional

cardiac muscle construct. J. Heart Lung Transplant. 24, 1091-1097 (2005).

122. C. Fink, S. Ergun, D. Kralisch, U. Remmers, J. Weil, and T. Eschenhagen, Chronic

stretch of engineered heart tissue induces hypertrophy and functional improvement.

FASEB J. 14, 669-679 (2000).

123. M. Papadaki, N. Bursac, R. Langer, J. Merok, G. Vunjak-Novakovic, and L. E. Freed,

Tissue engineering of functional cardiac muscle: molecular, structural, and

electrophysiological studies. Am. J. Physiol Heart Circ. Physiol 280, H168-H178 (2001).

124. M. Radisic, H. Park, H. Shing, T. Consi, F. J. Schoen, R. Langer, L. E. Freed, and G.

Vunjak-Novakovic, Functional assembly of engineered myocardium by electrical

Page 58: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

46

stimulation of cardiac myocytes cultured on scaffolds. Proc. Natl. Acad. Sci. U. S. A 101,

18129-18134 (2004).

125. J. Folkman and Y. Shing, Angiogenesis. J. Biol. Chem. 267, 10931-10934 (1992).

126. M. W. Laschke, A. Elitzsch, B. Vollmar, P. Vajkoczy, and M. D. Menger, Combined

inhibition of vascular endothelial growth factor (VEGF), fibroblast growth factor and

platelet-derived growth factor, but not inhibition of VEGF alone, effectively suppresses

angiogenesis and vessel maturation in endometriotic lesions. Hum. Reprod. (2005).

127. D. G. Stupack and D. A. Cheresh, Integrins and angiogenesis. Curr. Top. Dev. Biol. 64,

207-238 (2004).

128. R. K. Li, Z. Q. Jia, R. D. Weisel, D. A. Mickle, A. Choi, and T. M. Yau, Survival and

function of bioengineered cardiac grafts. Circulation 100, II63-II69 (1999).

129. T. Eschenhagen, C. Fink, U. Remmers, H. Scholz, J. Wattchow, J. Weil, W.

Zimmermann, H. H. Dohmen, H. Schafer, N. Bishopric, T. Wakatsuki, and E. L. Elson,

Three-dimensional reconstitution of embryonic cardiomyocytes in a collagen matrix: a

new heart muscle model system. FASEB J. 11, 683-694 (1997).

130. L. L. Chiu and M. Radisic, Scaffolds with covalently immobilized VEGF and

Angiopoietin-1 for vascularization of engineered tissues. Biomaterials 31, 226-241

(2010).

131. G. E. Davis, K. J. Bayless, and A. Mavila, Molecular basis of endothelial cell

morphogenesis in three-dimensional extracellular matrices. Anat. Rec. 268, 252-275

(2002).

132. L. D. Shea, E. Smiley, J. Bonadio, and D. J. Mooney, DNA delivery from polymer

matrices for tissue engineering. Nat. Biotechnol. 17, 551-554 (1999).

Page 59: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

47

133. W. H. Zimmermann, M. Didie, G. H. Wasmeier, U. Nixdorff, A. Hess, I. Melnychenko,

O. Boy, W. L. Neuhuber, M. Weyand, and T. Eschenhagen, Cardiac grafting of

engineered heart tissue in syngenic rats. Circulation 106, I151-I157 (2002).

134. T. Dvir, A. Kedem, E. Ruvinov, O. Levy, I. Freeman, N. Landa, R. Holbova, M. S.

Feinberg, S. Dror, Y. Etzion, J. Leor, and S. Cohen, Prevascularization of cardiac patch

on the omentum improves its therapeutic outcome. Proc. Natl. Acad. Sci. U. S. A 106,

14990-14995 (2009).

135. M. N. Giraud, E. Ayuni, S. Cook, M. Siepe, T. P. Carrel, and H. T. Tevaearai, Hydrogel-

based engineered skeletal muscle grafts normalize heart function early after myocardial

infarction. Artif. Organs 32, 692-700 (2008).

136. A. Lesman, M. Habib, O. Caspi, A. Gepstein, G. Arbel, S. Levenberg, and L. Gepstein,

Transplantation of a tissue-engineered human vascularized cardiac muscle. Tissue Eng

Part A 16, 115-125 (2010).

137. K. R. Stevens, K. L. Kreutziger, S. K. Dupras, F. S. Korte, M. Regnier, V. Muskheli, M.

B. Nourse, K. Bendixen, H. Reinecke, and C. E. Murry, Physiological function and

transplantation of scaffold-free and vascularized human cardiac muscle tissue. Proc.

Natl. Acad. Sci. U. S. A 106, 16568-16573 (2009).

138. K. Mueller-Stahl, T. Kofidis, P. Akhyari, D. H. Lee, A. Lenz, E. C. Martinez, F. Woitek,

and A. Haverich, Determinants of bioartificial myocardial graft survival and engraftment

in vivo. J. Heart Lung Transplant. 27, 1242-1250 (2008).

139. J. M. Kelm, V. Djonov, S. P. Hoerstrup, C. I. Guenter, L. M. Ittner, F. Greve, A.

Hierlemann, C. D. Sanchez-Bustamante, J. C. Perriard, E. Ehler, and M. Fussenegger,

Tissue-transplant fusion and vascularization of myocardial microtissues and

macrotissues implanted into chicken embryos and rats. Tissue Eng 12, 2541-2553

(2006).

140. W. H. Zimmermann, I. Melnychenko, G. Wasmeier, M. Didie, H. Naito, U. Nixdorff, A.

Hess, L. Budinsky, K. Brune, B. Michaelis, S. Dhein, A. Schwoerer, H. Ehmke, and T.

Page 60: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

48

Eschenhagen, Engineered heart tissue grafts improve systolic and diastolic function in

infarcted rat hearts. Nat. Med. 12, 452-458 (2006).

141. P. Zhang, H. Zhang, H. Wang, Y. Wei, and S. Hu, Artificial matrix helps neonatal

cardiomyocytes restore injured myocardium in rats. Artif. Organs 30, 86-93 (2006).

142. H. Sekine, T. Shimizu, K. Hobo, S. Sekiya, J. Yang, M. Yamato, H. Kurosawa, E.

Kobayashi, and T. Okano, Endothelial cell coculture within tissue-engineered

cardiomyocyte sheets enhances neovascularization and improves cardiac function of

ischemic hearts. Circulation 118, S145-S152 (2008).

Page 61: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

49

Chapter 3: A modular approach to cardiac tissue engineering Leung BM and Sefton MV Tissue Engineering Part A, 2010 June 29 PMID: 20504074

3.1 Abstract

Functional cardiac tissue was prepared using a modular tissue engineering approach with

the goal of creating vascularized tissue. Rat aortic endothelial cells (RAEC) were seeded onto

sub-millimetre size modules made of type I bovine collagen supplemented with MatrigelTM

(25% v/v) embedded with cardiomyocyte (CM)-enriched neonatal rat heart cells and assembled

into a contractile, macroporous sheet-like construct. Modules (without RAEC) cultured in 10%

bovine serum were more contractile and responsive to external stimulus (lower excitation

threshold, higher maximum capture rate and greater en face fractional area changes) than

modules cultured in 10% fetal bovine serum. Incorporating 25% Matrigel in the matrix reduced

the excitation threshold and increased the fractional area change relative to collagen only

modules (without RAEC). A co-culture medium, containing 10% bovine serum, low Mg2+

(0.814 mM) and normal glucose (5.5 mM) was used to maintain RAEC junction morphology

(VE-cadherin) and CM contractility, although the responsiveness of CM was attenuated with

RAEC on the modules. Macroporous, sheet-like module constructs were assembled by partially

immobilizing a layer of modules in alginate gel until day 8, with or without RAEC. RAEC/CM

module sheets were electrically responsive, however, like modules with RAEC this

responsiveness was attenuated relative to CM only sheets. Muscle bundles co-expressing cardiac

troponin I and connexin 43 were evident near the perimeter of modules and at inter-module

junctions. These results suggest the potential of the modular approach as a platform for building

vascularised cardiac tissue.

Page 62: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

50

3.2 Introduction

Congestive heart failure (CHF) is a major cause of death in both the developed and

developing world. Although the pathophysiologies relating to CHF are complex, the condition is

ultimately caused by the weakening of heart muscle tissue resulting in lowered cardiac output.

The decrease in cardiac output causes chronic systemic ischemia that leads to decompensation

responses such as ventricular hypertrophy, increased blood volume, and increased blood

pressure, which in turn increases cardiac load and leads to further damage to cardiac muscle

tissue. As a result, there is considerable interest in fabricating a cardiac tissue substitute using

cardiomyocytes in vitro. The resulting tissue engineered cardiac muscle would be expected, at a

minimum, to possess similar mechanical and functional properties as native cardiac tissue, but

also have the capability to be integrated into native tissue thus preventing long term cardiac

tissue remodeling1.

Attempts to fabricate functional cardiac tissues in vitro have yielded promising results2,3.

A crucial factor in creating viable 3-dimensional tissue in vitro is to achieve adequate perfusion

throughout the tissue. This is especially true for cardiac tissue due to its high metabolic rate and

oxygen demand. With a view to improving engineered tissue perfusion, Radisic et al.4

incorporated oxygen carriers and flow channels into neonatal rat cardiomyocyte (CM) seeded

scaffolds. Aside from nutrients, environmental factors including mechanical5 and electrical6

stimuli are also needed for the proper development of cardiac tissue. Many of these concepts

have been incorporated into recent culture systems7 in which CM are incorporated with other

materials to form functional tissues 8. For example, a 3-dimensional model was used by

Zimmermann et al.5, where they embedded a neonatal rat CM enriched cell population (also

containing fibroblasts and endothelial cells, EC) into a MatrigelTM/collagen gel. In their model,

the EC in culture facilitated angiogenesis and host vasculature coupling. Other 3-dimensional

culture systems rely on cell secreted extracellular matrix (ECM) as the major matrix component.

Kelm et al.9,10 created microtissues containing CM in a hanging drop culture system. The scale

of these microtissues (~100 µm) has been shown to support core tissue viability. In addition,

contractile CM sheets have been cultivated and assembled into multilayer tissues by Shinizu et

al.11. These strategies improve tissue perfusion by promoting either angiogenesis of surrounding

blood vessels, or neovasculogenesis of embedded endothelial cells (part of the CM mixed cell

population) via cytokine and chemokine signaling.

Page 63: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

51

The overall goal of this work is to create functional, cardiac tissue using a modular tissue

engineering approach with neonatal rat CM embedded in a collagen/MatrigelTM gel (the module)

and seeded with rat aortic endothelial cells (RAEC). Modules had a post-contraction length of

approximately 500 µm and diameter of 300 µm. When assembled into a construct, the resulting

interconnected channels are perfuseable, mimicking the required vasculature. The small

diameter of each module ensures sufficient nutrient delivery to the centre by diffusion. Modules

may be assembled into larger constructs in vitro followed by implantation, or they may be

deployed in vivo and allowed to be remodeled by host tissue. We hypothesized that the modular

tissue engineering approach 12,13 can be used to produce vascularized cardiac tissue by

assembling modules into a macroporous sheet, where EC in the intermodular space can form

vessel-like structures. This approach results in uniform, scalable and vascularized constructs14

and is being explored elsewhere for its utility in pancreatic islet transplantation and fat

reconstruction.

In work reported here, we have characterized the in vitro phenotype and functional

response of a CM/RAEC co-culture system comprised of both individual modules and modules

in the form of a contractile sheet. We examined the responses of CM-only and RAEC/CM co-

culture modules to external electrical stimulation, as well as the effect of surface coated RAEC

on selected features of the embedded CM. Key aspects of this study was the definition of a

suitable medium for co-culture of CM and EC and a means of preparing sheet-like structures

from the modules.

Page 64: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

52

3.3 Materials and Methods

3.3.1 Cardiomyocyte isolation and module fabrication

The protocol for cell isolation was similar to that reported by Radisic et al6. Hearts from

neonatal rat pups (1-2 days old) were harvested and kept in ice cold HBSS (Hanks balanced salt

solution, Gibco, Mississauga, ON). The aortas were trimmed and the remaining heart tissues

were quartered followed by serial digestion with bovine pancreas trypsin (0.6mg/mL in HBSS,

Sigma, Mississauga, ON) and collagenase II (1 mg/mL in HBSS, Worthington Biochemical,

Lakewood, NJ). The extracted cell mixture was pre-plated onto tissue culture polystyrene for 1

hour to enrich its CM content. Harvested cells were maintained in native CM medium consisting

of high glucose (25 mM) DMEM medium (Gibco) supplemented with 10% fetal bovine serum

(FBS; or 10% bovine serum, BS, Sigma), 1% HEPES buffer (10 mM, Gibco), and 1%

penicillin/streptomycin (Gibco). Each CM preparation consisted of cells from an entire litter (10

to 13 neonatal rats) to enhance consistency. Over the course of the study several litters were

used in each set of data. The number of experiments reflects the use of these replicate litters.

The CM enriched cell mixture was loaded onto a Cytospin® slide and stained for

troponin I and prolyl-4-hydroxylase (see below) to identify CM and fibroblasts respectively: the

mixture consisted of 50% cardiomyocytes and 40% fibroblasts. The remaining 10% was

presumed to consist of other cardiac cell types, including EC. The CM-rich cell mixture was

embedded in a type I bovine collagen matrix supplemented with 25% MatrigelTM (v/v, BD

Bioscience), and cast into modules using methods as described previously 15. Viability of

embedded cells was determined using Live/DeadTM assay kit (Molecular Probe, Eugene, OR).

Page 65: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

53

3.3.2 Endothelial cell seeding

RAEC were purchased and cultured in an MCDB-131 based native EC medium (VEC

Technologies, Rensselaer, NY), supplemented with 10% FBS. In some experiment, RAEC were

transduced with a lentiviral eGFP vector16 to enable tracking over time (in collaboration with

Dr. J. Medin, Ontario Cancer Institute).

CM-embedded modules were cultured in native CM medium supplemented with 10%

BS for 3 days prior to EC seeding. To coat the surface of modules, 2-3 million RAEC were

suspended in a seeding medium consisting of a 50:50 mixture of native CM (10% BS) and

native EC (10% FBS) medium, and mixed with one mL of CM-embedded modules inside a 15

mL centrifuge tube. The tube was agitated for 30 minutes on a uniaxial rocker at low speed. The

mixture was transferred to a non-tissue culture treated 6-well plate and kept in seeding medium

overnight in order to promote RAEC attachment. The seeding medium was used to transition the

CM and EC from their separate native media to the co-culture medium, described in Table 3-1.

After one day, modules were transferred into a new non-tissue culture treated 6-well plate to

remove any RAEC that did not attach to modules, and the seeding medium was changed to the

co-culture medium. Modules were maintained in this medium until assayed.

Table 3-1: Medium compositions (presumed key differences) and serum types used in the

study.

Page 66: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

54

3.3.3 Module sheet

Four days after fabrication, CM embedded modules were assembled into a monolayer

sheet. For RAEC/CM modular sheets, CM embedded modules (3 days after fabrication) were

seeded with RAEC and assembled into sheets the next day. Using a 10 mL serological pipette,

modules, with or without RAEC, were placed over a sterile nylon mesh (pore size = 100 µm,

Millipore, Cork, Ireland) to form a single layer. A dilute alginate solution (1.2% w/w, Sigma)

was dripped over the modular sheet followed by a few drops of 50 µM calcium chloride to

crosslink the alginate. Once the alginate formed a soft gel (approximately 5 to 10 seconds), the

sheet was rinsed in PBS (1.06 mM KH2PO4, 155.17 mM NaCl, 2.97 mM Na2HPO4-7H2O) and

transferred into CM or co-culture medium. These alginate embedded sheets were stimulated (see

below) for 10 days and then the alginate gel was removed prior to assay by brief incubation in

100 µM citrate buffer at room temperature.

3.3.4 Electrical response assessment and field stimulation

CM embedded modules were equilibrated in 1x Tyrode’s salt solution (Sigma) at 37oC

for 1 hour prior to measurement. Electrical responses of modules were assessed using custom

made testing chambers. Modules were paced between a pair of graphite electrodes kept 1 cm

apart by a polycarbonate bracket. Using a 6-well plate made from tissue culture polystyrene,

modules and electrodes were placed in an individual well with 1.5 mL of Tyrode’s salt solution,

which was enough to partially submerge the electrodes. The small chamber volume ensured that

most of the modules were located between the electrodes.

Module sheets were assayed using a PDMS/glass testing chamber. A pair of graphite

electrodes were kept 1 cm apart by a PDMS bracket and placed over a layer of PDMS poured

into a 60mm glass Petri dish. This allowed the module sheets to be fixed in position relative to

the electrode.

For both systems, a signal generator (S88X stimulator, Grass Technologies, Brossard,

QC) connected to the electrodes provided the desired electrical stimulation (biphasic, 2 ms

square pulse, up to 12 V at various frequencies). The samples were kept at 37oC during the

experiment using a solid-state microscope stage heater. Contractions of modules and module

sheets were recorded with a CCD camera (Hitachi Denshi, model KP-M1U) on an inverted

Page 67: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

55

phase contrast microscope (Olympus CX2). The image sequences were analyzed using ImageJ

(NIH, version 1.38x).

Excitation threshold (ET) was defined as the minimum voltage required for synchronous

contraction at 1 Hz. Maximum capture rate (MCR) was defined as the maximum rate of

synchronous contraction under an electric field equivalent to twice the ET. The contractility of

modules was defined as the fractional en face area change during one contraction cycle using

frame-by-frame video analysis in ImageJ.

Module sheets were electrically conditioned prior to assay. Briefly, CM-embedded

modules were maintained in native CM medium with 10% BS for 3 days. Then the modules

were fabricated into sheets (see above) and transferred into a well, fitted with a pair of graphite

electrodes as above, and stimulated continuously with biphasic pulses at 5 V/cm and 1 Hz for 10

days. Medium was supplemented with 10 µM of ascorbic acid and changed daily to minimize

the effect of reactive oxygen species generated at the electrodes. Individual modules were not

electrically conditioned because they could not be fixed in position relative to the electrical field

over the course of an experiment.

3.3.5 Immunofluorescence staining

Freshly harvested, CM enriched cell mixtures were analyzed by cytospin. Slides were

stained with rabbit anti-rat troponin I (Santa Cruz Biotechnology, Santa Cruz, CA) and mouse

anti-rat prolyl 4-hydroxylase (Sigma, Oakville, ON) antibodies to identify cardiomyocytes and

fibroblasts, respectively.

RAEC coverage on modules was determined by whole mount immunoflourescence

staining of goat anti-rat VE-cadherin primary antibody (Santa Cruz Biotechnology, Santa Cruz,

CA) followed by rabbit anti-goat IgG AlexaFluorTM 488 secondary antibody (Molecular Probes,

Eugene, OR). Modules were fixed in 4% paraformaldehyde solution and permeabilized using

0.2% Triton-X100 solution. On immunohistochemical sections, CMs were stained using rabbit

anti-rat troponin I (Santa Cruz Biotechnology, Santa Cruz, CA) followed by goat anti-rabbit IgG

secondary antibodies conjugated with AlexaFluorTM 488 (Molecular Probes, Eugene, OR).

Cardiac fibroblasts were stained with mouse anti-rat vimentin antibody (Sigma, Oakville, ON)

followed by goat anti-mouse IgG AlexaFluor 568 (Molecular Probes, Eugene, OR). In sections

that were triple stained (troponin I, vimentin and connexin-43), connexin-43 was stained prior to

immunofluoresence staining using rabbit anti-rat Cx43 antibody (Chemicon) coupled by a three-

Page 68: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

56

step IHC chromogeninc method. In some experiments, surface seeded RAEC were tracked by

eGFP, where cell nuclei were counterstained with Hoechst nuclear stain (Sigma, Oakville, ON).

Samples were visualized using a Zeiss LSM510 confocal microscope.

3.3.6 Statistical analysis

Statistical analysis including calculations of standard error of means and mean

comparisons (t-test or one-way ANOVA) were performed using Prism (Version 5.0, Graphpad

Software, La Jolla, CA)

Page 69: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

57

3.4 Results

3.4.1 Contractility of CM embedded modules

Cardiomyocytes embedded in modules remained viable for at least 14 days in either type

1 collagen or type 1 collagen supplemented with 25% MatrigelTM. However, higher viability

was obtained with an initial cell seeding of 107 cells/mL compared to 106 cells/mL (Figure 3-1).

Modules without RAEC were cultured for 7 days in cardiomyocyte medium containing either

10% FBS or 10% BS and then assayed for electrical responsiveness. In both cases, modules

displayed synchronous contractions corresponding to the external stimulation frequency (Figure

3-2). A small proportion of the modules (< 10%) contracted spontaneously even in the absence

of external electrical stimulation and continued to contract when stimulated. Modules cultured

in 10% BS had a lower excitation threshold but similar maximum capture rate compared to

modules cultured in 10% FBS (Figure 3-3A). Moreover, modules cultured in 10% BS had better

defined peaks with higher maximum contraction amplitudes, compared to modules cultured in

10% FBS; a representative sequence of a single module (from a group of >30) is shown in

Figure 3-3B. Histological sections of modules cultured in 10% BS showed higher troponin I

expression and elongated cell morphology compared to modules cultured in 10% FBS (Figure

3-3C). These results suggest that bovine serum enhanced the electrical responsiveness and

contractile performance of CM-embedded modules, through an effect on the expression or

organization of the contractile apparatus within the cells. For this reason, all subsequent

experiments were carried out in 10% BS supplemented medium.

The effect of extracellular matrix on the function of embedded CM (10% BS

supplemented medium) was tested by embedding CM in either collagen (type 1) or collagen

supplemented with MatrigelTM; the latter choice was based on the work of Zimmerman et al5.

CM embedded in MatrigelTM supplemented matrix exhibited lower excitation thresholds (Figure

3-4A, p<0.05) and higher contractile amplitudes (Figure 3-4C) compared to CM embedded in

collagen only modules. However, the maximum capture rates were found to be similar for the

two groups (Figure 3-4B).

Page 70: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

58

Figure 3-1: Viability (live/dead assay) of cardiomyocytes embedded in modules, cultured in

10% BS, at 14 days post fabrication. Matrix composition did not affect embedded cell viability,

while increasing seeding density from 106 to 107 cell/mL dramatically improved viability.

(Magnification = 100x)

Page 71: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

59

Figure 3-2: Fractional area change of a single module when stimulated by external signal. CM-

embedded modules (2 x 107 cell/mL collagen supplemented with 25% v/v MatrigelTM) were

cultured in 10% BS medium for 7 days prior to assay. At a voltage twice the excitation

threshold, modules displayed synchronous contraction corresponding to the external signal

frequency: 1 Hz from 0 to 7 sec, 2 Hz from 7 to 13 sec, and 3 Hz from 13 to 17 sec.

Page 72: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

60

Figure 3-3: (A) Excitation thresholds (ET) and maximum capture rates (MCR) of CM (2 x 107

cell/mL) modules (collagen with 25% MatrigelTM) cultured in medium containing 10% FBS

(solid) or 10% BS (hatched) for 7 days post fabrication. Maintaining modules in medium with

10% BS decreased ET (*, p<0.05, n=3), but did not affect MCR. (B) Fractional area change of

single modules (taken from an aliquot of >30) on day 7. Modules cultured in 10% BS or 10%

Page 73: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

61

FBS were stimulated at 200% excitation threshold voltage (2ms, 1 Hz) and recorded on video.

Fractional area change is normalized against a reference frame in each sample. (C) CM cultured

in 10% BS expressed more cardiac troponin I (green) and assumed a more elongated

morphology than those cultured in 10% FBS when examined using immunofluorescent

microscopy.

Figure 3-4: At day 7 post fabrication, CM (2 x 107 cell/mL) embedded in MatrigelTM (25% v/v)

supplemented collagen modules, cultured in 10% BS, exhibited a lower excitation threshold (A,

*, p<0.05, n=3) and higher contractility (C, single modules taken from an aliquot of >30)

compared to collagen only control. However, both collagen only and collagen+MatrigelTM

modules had similar maximum capture rates (B).

Page 74: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

62

3.4.2 Behavior of RAEC seeded CM embedded modules

Primary RAEC (2 x 107 cell/mL) were seeded onto modules embedded with CM 3 days

after fabrication to form endothelialized modules; approximately 25% of the module surface

was covered at initial seeding. After 24 hours in a seeding medium consisting of a 50:50 mixture

of the two native media, the RAEC/CM modules were placed in a co-culture medium that had a

lower glucose concentration (5.5 mM) than the native CM medium and a much lower Mg2+

concentration than native EC medium (Table 3-1). These factors, among other perhaps less

crucial medium differences, helped maintain the contractility of embedded CM and promoted

EC adherent junction formation. After 14 days in co-culture medium, the surface of the modules

were almost completely covered by RAEC with well-developed adherent junctions, confirmed

by junction localized VE-cadherin staining (Figure 3-5A), similar to that seen with RAEC

seeded on empty collagen modules and cultured in native EC medium. In contrast, RAEC

seeded on modules cultured in native cardiomyocyte medium showed no morphologically

distinguishable junction features (Figure 3-5B).

When stimulated by an external electric field, endothelialized modules were able to

contract synchronously, but they had a higher excitation threshold (p<0.001) and lower

maximum capture rate (p<0.05) compared to CM-only modules (Figure 3-5C). In fact their

electrical response profile resembled that of CM-only modules cultured in 10% FBS. However,

both endothelialized modules and CM-only modules had similar contraction amplitudes (Figure

3-5D).

Page 75: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

63

Figure 3-5: (A) Immunofluorescence staining for VE-Cadherin on RAEC/CM modules after 14

days in co-culture medium indicated the presence of a confluent layer of RAEC. (B) In contrast,

RAEC seeded on CM modules cultured in high glucose native CM medium displayed

disorganized junctions. (C) Surface coated RAEC attenuated the electrical responsiveness of

modules, as seen by an increase in excitation threshold (***, p<0.0001, n=3) and decrease in

maximum capture rate (*, p<0.05, n=3). (D) With external field stimulation, CM-embedded

modules coated with RAEC displayed similar contractility when compared to CM-only modules

(single modules taken from an aliquot of >30). Contractility was assessed by comparing the en

face fractional area change.

Page 76: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

64

3.4.3 Module sheets

The modular approach mimics the multilayered organization of cardiac muscle by first

fusing sub-millimeter modules into a sheet-like construct with a view to subsequently stacking

these “sheets” together to yield a perfuseable, macroporous, endothelialized three dimensional

tissue. One of the challenges of assembling multiple cell embedded modules into a sheet is to

control the rate and degree of agglomeration caused by remodeling. Modules embedded with

contractile cell types, such as smooth muscle cells17 and CM, tend to clump and fuse into a large

cell mass spontaneously under static conditions. The size of these non-porous aggregates is

expected to impair diffusion and ultimately lead to necrosis of the core. To minimize this, the

modules were partially embedded in a thin layer of alginate gel, a non cell adhering matrix, to

prevent the spaces between modules from collapsing, while at the same time allowing a limited

amount of intermodule fusion to occur (Figure 3-6A). In as little as five days, a macroporous

module sheet was produced, and then the alginate was dissolved, releasing the fragile, yet

physically stable sheet from the supporting nylon mesh (Figure 3-6B).

Both CM only as well as RAEC/CM module sheets were characterized. At 4 days post

fabrication, modules were assembled into sheets and stimulated for an additional 10 days.

RAEC/CM modules were made by seeding eGFP tranduced RAEC onto modules one day prior

to sheet assembly (i.e., on day 3). After 10 days of stimulation, RAEC were tracked and found

to be evenly distributed over the surface of module sheets (Figure 3-6D). Moreover, the

attachment density was enhanced by removing the alginate gel and reseeding the sheet with

more RAEC (Figure 3-6E), after 10 days of stimulation. When placed in an electrical field

(Figure 3-6C), both RAEC/CM and CM-only module sheets contracted synchronously.

RAEC/CM sheets exhibited a higher excitation threshold (p<0.01), but a roughly similar

maximum capture rate compared to CM-only sheets (Figure 3-6C). When RAEC/CM sheets

were not stimulated for the 10 days of culture, they did not contract when placed in an electric

field.

Module sheets with or without RAEC were stained for cardiac troponin I, a protein

subunit associated with the troponin-tropomyosin complex in cardiac muscle. Troponin I

positive cells were found throughout the modules. However, most of the muscle bundle-like

structures were found at the perimeter of the modules or near the inter-module junctions (Figure

3-7). Most of these structures were no more than 5 cell layers thick and covered 12 ± 2% (n = 4)

Page 77: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

65

of the cross sectional area in both RAEC/CM or CM-only sheets (Figure 3-7B). When triple-

stained with vimentin, troponin I and connexin-43, we saw a fibroblast rich layer at the

periphery of the modules (Figure 3-8A). We also found connexin-43 co-localized with troponin

I positive structures along the perimeter of modules and inter-modular space, just below the

fibroblast layer (Figure 3-8C, D).

Page 78: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

66

Figure 3-6A: (A) Schematic diagram of the testing chamber used to condition module sheets.

Modules were partially embedded in alginate gel on top of a nylon mesh and conditioned

electrically for 10 days.

Page 79: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

67

Figure 3-6(B-E): (B) After 10 days of continuous stimulation, macroporous RAEC-CM module

sheets were removed from the alginate and nylon mesh and assessed by field stimulation. (C)

Excitation thresholds and maximum capture rates of RAEC/CM sheets and CM-only sheets;

excitation threshold was significantly higher in the presence of RAEC (**, p<0.001, n=3). (D)

eGFP transfected RAEC were uniformly distributed, at a relatively low density, over RAEC/CM

modular sheets, 10 days after stimulation. (E), RAEC/CM sheets were reseeded with 3 x106

RAEC, using the same protocol as in the first seeding, after removing the alginate gel. With this

protocol, RAEC surface coverage was increased.

Page 80: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

68

Figure 3-7: (A) Immunofluorescence staining of cardiac troponin I (green) shows a high density

of CM near the perimeter of modules and at inter-module junctions (dash lines indicate edges of

modules) in a module sheet after 10 days of stimulation. (B) At higher magnification, the CM

appeared to form muscle like bundles, similar to that seen in native cardiac tissue, but such

bundles constituted only ~12% of the field, shown in A.

Page 81: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

69

Figure 3-8: (A-B) Triple-stained (vimentin, troponin I and connexin-43) modular sheets at after

10 days of stimulation indicate that surfaces of module sheets were covered by a layer of

fibroblasts with an underlying layer of CM. (C-D) Connexin-43 positive structures were also

observed. (E-F) Co-localization of connexin-43 and troponin I was confirmed by overlaying

high magnification images suggesting the CM were electrically coupled.

Page 82: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

70

3.5 Discussion

We have characterized sub-millimeter sized modules containing cardiomyocytes and

endothelial cells and examined the feasibility of building larger constructs with this modular

approach. We focus here on characterizing the individual modules and the assembled sheet-like

structures. Future reports will describe the fate of these modules and sheets in cardiac infarct

models.

3.5.1 Electrical characterization

CM-embedded modules responded to external field stimulation after 7 days in culture.

Addition of MatrigelTM (25% v/v) to the collagen matrix lowered the excitation threshold and

improved the contractility of the modules, but did not affect the maximum capture rate. This

benefit is attributed to the presence of basal lamina proteins that are found in MatrigelTM,

including laminin and collagen type IV, as well as the various growth factors that are found in

MartigelTM. The presence of MatrigelTM is presumed to provide an extracellular matrix

environment that is more similar to the native myocardium than collagen type I alone, resulting

in improved cell organization and tissue remodeling6. For further development of this approach,

an alternative is required since the tumour derived Matrigel has limited utility in clinical

applications.

Culturing modules in 10% BS instead of 10% FBS also resulted in improved electrical

performance and contractility, and this was accompanied by an increase in troponin I expression

and cell elongation. This observation is consistent with that of Schwarzkopf et al.18 who showed

that medium supplemented with adult serum (same species) promoted viability and maturation

of cardiomyocytes compared to fetal calf serum. While we did not investigate the effect of adult

rat serum on rat CM, we saw a similar improvement in CM maturation with adult bovine serum

in comparison to fetal bovine serum. It was beyond the scope of this study to elucidate the

underlying mechanism. Similarly, further optimizations of the matrix or the medium are left for

the future although the value of this for rat CM or EC may not be warranted.

We acknowledge that the current method for assessing electrical performance is highly

dependent on the materials used and the geometry of the testing chamber, and therefore cannot

be viewed as a true analog for representing in vivo electrical performance. Of particular

Page 83: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

71

importance is the inability to assess regional differences in contractility and whether action

potentials propagated throughout the thickness of the construct or just merely on the surface.

Nevertheless, the current method allowed us to compare the overall effects exerted by different

environmental signals and enabled us to optimum culture conditions.

3.5.2 Co-culture model

One of the main objectives of this study was to develop a means of maintaining a

functional RAEC layer on the surface of contractile CM-embedded modules. By adjusting the

composition of the medium (Table 1) and lowering the glucose level to that of native EC

medium, primary RAEC remained viable and showed typical VE-cadherin expression after 14

days, suggesting the endothelial cells were in a quiescent state19 in the co-culture medium.

Using the same medium we were also able to generate viable RAEC/CM co-culture modular

sheets.

To our knowledge, this is the first study to investigate the effects of surface coated

endothelial cells on the electrical performance and responsiveness of underlying CM in a

modular construct. A 24 hour transition period and seeding medium (a 50:50 mixture of the

native media) was used to enable RAEC seeding and coverage. Without this seeding medium,

RAEC attachment and coverage was poor. We found that the presence of a quiescent RAEC

layer in the co-culture medium, on either modules or modular sheets, permitted the contractile

response of embedded CM, provided the Mg2+ concentration was at the low level of CM

medium and not the high value of the native EC medium. However, the RAEC reduced the

module and module sheet electrical responsiveness, manifested as an increase in excitation

threshold and decrease in maximum capture rate. These changes in electrical performance were

not seen when CM-only modules were cultured in the same co-culture medium. We believe that

these changes were most likely caused by the presence of the RAEC layer which may have

changed the surface conductance of the modules. Such changes would cause the excitation

threshold to increase compared to CM-only modules. Differences in membrane protein

expression (e.g. connexin proteins), may also attenuate the propagation of electrical impulses

from the surface to the underlying CM, resulting in a lower maximum capture rate.

Judging from VE-cadherin staining, the native cardiac EC embedded in the modules did

not form vessel-like structures, nor did they appear to interact with surface seeded RAEC.

Page 84: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

72

However, these cardiac EC may still play an important role in CM maturation, as demonstrated

in other multi-culture cardiac constructs20-22. For example, Iyer et al. showed the presence of

fibroblasts and EC in cardiac tissue constructs enhanced CM elongation and excitability23,

possibly by spatially orienting CM into muscle-like bundles.

Our initial aim was to use the RAEC/CM modules as a vehicle to uniformly deliver

viable EC through out an assembled construct containing multiple modules. In a separate study

involving RAEC modules (without embedded cells) implanted in a rat omental pouch, we

observed that RAEC migrate from the modules and form chimeric (host-donor vessels)

functional blood vessels in the intermodular space after 21 days24. We expect the RAEC on the

co-culture modules to migrate and form vessels in a similar fashion, when implanted. Plans to

evaluate the modular construct in a rat implant model are expected to reveal the fate and

remodeling process of RAEC/CM modules in vivo; how this then changes the in vivo electrical

properties and morphology of CM remains to be defined.

3.5.3 Sheet formation

A major challenge in creating clinically relevant, thick (1 to 10 mm) cardiac tissues in

vitro is the lack of tissue vasculature. Specifically, the formation of blood vessels, both host and

graft derived, can be quite slow and insufficient to meet the high nutrient demands of

cardiomyocytes. There are several approaches designed to circumvent this limitation, including

the use of oxygen carriers in culture medium25, incorporating vessel-like flow channels in

preformed scaffolds26, and preloading scaffolds with various growth factors and/or functional

cell types that promotes angiogenesis27,28.

We devised a method to produce a sheet-like structure from an assemblage of modules.

A thin layer of modules was laid onto a nylon mesh and partially immobilized in alginate gel. At

least some of the CM in these modules matured into muscle-like bundles and formed a

physically integrated sheet capable of synchronous contraction when stimulated. Moreover, the

CM at inter-modular spaces expressed connexin-43, suggesting that the module sheet was

electrically coupled. However, these muscle-like bundles, as observed on histological sections,

are relatively thin and only occupy a fraction of the total volume of the module. The lack of

troponin I positive CM near the core of the module suggests that the CM may have been

crowded out by the faster growing fibroblasts. However this may also be the result of active CM

Page 85: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

73

migration and maturation. Further studies at different time points are needed to reveal the

mechanism of the remodeling process. Since we assume that these bundles are responsible for

generating the contractile forces, it is crucial to increase the proportion of these bundles for the

modular construct to produce physiologically relevant force. For instance, in future

implementations it may be beneficial to reduce the diameter of modules so that the surface to

volume ratio is increased, thus increasing the contractile force on a per module basis. It is also

conceivable that over a longer incubation period in vitro or when implanted in vivo, more CM

may migrate to the surface thus increasing the thickness of this layer. These issues will be

explored in future studies.

In recent years, there has been interest in building cardiac constructs using small

functional sub-units: e.g., micro-tissues9, cardiac sheets11 or other components29. In the approach

most conceptually similar to modules, Kelm et al. demonstrated that human umbilical vein

endothelial cell (HUVEC) coated micro-tissue (a “spheroid” without collagen gel) with

embedded CM were able to agglomerate to form a patch with functional HUVEC lined

capillaries10,30. In all cases, the goal is to create uniform, scalable, vascularized and functional

cardiac tissue and the merits of one approach relative to the other will be determined by the

success of meeting these goals based on in vivo performance. Of particular significance in this

work is that co-culture conditions that maintained both CM function and RAEC junction

morphology were devised. We found, for example, that RAEC/CM sheets that were not

stimulated during culture did not contract when subject to external field stimulation at the time

of analysis (after 10 days of stimulation).

We chose to embed modules in alginate not only because of its ease of application and

removal by changing local calcium concentration, but also because it is cell compatible and non

cell adherent. A single-module thickness across the sheet ensured that sufficient nutrients could

diffuse into the core of each module, at least in vitro. These macroporous module sheets will

serve as intermediate components for eventually building thicker tissues in a scalable fashion,

where the tissue maturation process can be monitored. When the modules were assembled into

sheets, there was a small decrease in the electrical responsiveness relative to modules alone.

This is likely the result of differing geometries of the stimulation apparatus resulting in

differences in the field perceived by the CM. It is also conceivable that intermodular interactions

of surface seeded RAEC may have lead to a change in tissue conductance.

Fabrication of functional cardiac tissue in vitro depends on many parameters, including

medium composition, extracellular matrix, geometry and cell seeding sequence. Native

Page 86: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

74

myocardium consists of cardiomyocytes that form mechanically and electrically coupled

bundles. These bundles are encased in ECM secreted by cardiac fibroblasts, and maintained by

capillaries lined with endothelial cells. The three cells types exist in their respective

microenvironments. The RAEC/CM co-culture system described in this study consists of more

than two cell types, since the primary CM enriched cell population contained a significant

quantity of fibroblasts and cardiac EC. While in this study, the electrical responsiveness of CM

and the adherent junction morphology of RAEC were chosen as the primary endpoints, we must

also consider the contribution of cardiac fibroblasts and EC to these measurements. The data

presented here suggest that cardiac fibroblasts actively segregate and migrate to the surface of

the modular sheet, and potentially play an important role in the formation and remodeling of the

modular sheet since the spatio-temporal relationships between CM and the various supporting

cell types has been shown to enhance CM maturation and the overall functionality of

construct23,31. Therefore, aside from being a viable platform for in vitro tissue formation, the

module system may also serve as a cell delivery vehicle that is capable of providing a supportive

environment to multiple cell types in a pre-determined seeding ratio and geometry.

3.6 Conclusion

We have created rat aortic endothelial cell coated, neonatal rat cardiomyocyte embedded

modules as well as module sheets. In both cases, embedded cells formed troponin I positive,

native muscle-like structures (albeit to a limited extent), and contracted with external field

stimulation. In the presence of endothelial cells in an appropriate co-culture medium, the

construct became less responsive, although it continued to be excitable and contractile. While an

in vivo study will be needed to elucidate the fate of these modules, this study has demonstrated

the feasibility of creating functional cardiac tissue using the modular approach.

Page 87: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

75

Reference List

1. D. Schmidt, A. Mol, S. Neuenschwander, C. Breymann, M. Gossi, G. Zund, M. Turina,

and S. P. Hoerstrup, Living patches engineered from human umbilical cord derived

fibroblasts and endothelial progenitor cells. Eur. J. Cardiothorac. Surg. 27, 795-800

(2005).

2. R. L. Carrier, M. Papadaki, M. Rupnick, F. J. Schoen, N. Bursac, R. Langer, L. E. Freed,

and G. Vunjak-Novakovic, Cardiac tissue engineering: cell seeding, cultivation

parameters, and tissue construct characterization. Biotechnol. Bioeng. 64, 580-589

(1999).

3. R. K. Li, Z. Q. Jia, R. D. Weisel, D. A. Mickle, A. Choi, and T. M. Yau, Survival and

function of bioengineered cardiac grafts. Circulation 100, II63-II69 (1999).

4. M. Radisic, H. Park, F. Chen, J. E. Salazar-Lazzaro, Y. Wang, R. Dennis, R. Langer, L.

E. Freed, and G. Vunjak-Novakovic, Biomimetic approach to cardiac tissue engineering:

oxygen carriers and channeled scaffolds. Tissue Eng 12, 2077-2091 (2006).

5. W. H. Zimmermann, C. Fink, D. Kralisch, U. Remmers, J. Weil, and T. Eschenhagen,

Three-dimensional engineered heart tissue from neonatal rat cardiac myocytes.

Biotechnol. Bioeng. 68, 106-114 (2000).

6. M. Radisic, H. Park, H. Shing, T. Consi, F. J. Schoen, R. Langer, L. E. Freed, and G.

Vunjak-Novakovic, Functional assembly of engineered myocardium by electrical

stimulation of cardiac myocytes cultured on scaffolds. Proc. Natl. Acad. Sci. U. S. A 101,

18129-18134 (2004).

7. W. L. Grayson, T. P. Martens, G. M. Eng, M. Radisic, and G. Vunjak-Novakovic,

Biomimetic approach to tissue engineering. Semin. Cell Dev. Biol. 20, 665-673 (2009).

8. G. Vunjak-Novakovic, N. Tandon, A. Godier, R. Maidhof, A. Marsano, T. Martens, and

M. Radisic, Challenges in Cardiac Tissue Engineering. Tissue Eng Part B Rev. (2009).

9. J. M. Kelm, E. Ehler, L. K. Nielsen, S. Schlatter, J. C. Perriard, and M. Fussenegger,

Design of artificial myocardial microtissues. Tissue Eng 10, 201-214 (2004).

Page 88: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

76

10. J. M. Kelm, V. Djonov, L. M. Ittner, D. Fluri, W. Born, S. P. Hoerstrup, and M.

Fussenegger, Design of custom-shaped vascularized tissues using microtissue spheroids

as minimal building units. Tissue Eng 12, 2151-2160 (2006).

11. T. Shimizu, M. Yamato, Y. Isoi, T. Akutsu, T. Setomaru, K. Abe, A. Kikuchi, M.

Umezu, and T. Okano, Fabrication of pulsatile cardiac tissue grafts using a novel 3-

dimensional cell sheet manipulation technique and temperature-responsive cell culture

surfaces. Circ. Res. 90, e40 (2002).

12. A.P.McGuigan and M.V.Sefton, Design and Fabrication of a vascularized tissue

engineered construct. Cardiovascular Pathology 13, S182 (2004).

13. A. P. McGuigan and M. V. Sefton, Vascularized organoid engineered by modular

assembly enables blood perfusion. Proc. Natl. Acad. Sci. U. S. A 103, 11461-11466

(2006).

14. A. P. McGuigan and M. V. Sefton, Vascularized organoid engineered by modular

assembly enables blood perfusion. Proc. Natl. Acad. Sci. U. S. A 103, 11461-11466

(2006).

15. A. P. McGuigan and M. V. Sefton, Vascularized organoid engineered by modular

assembly enables blood perfusion. Proc. Natl. Acad. Sci. U. S. A 103, 11461-11466

(2006).

16. M. Yoshimitsu, K. Higuchi, S. Ramsubir, T. Nonaka, V. I. Rasaiah, C. Siatskas, S. B.

Liang, G. J. Murray, R. O. Brady, and J. A. Medin, Efficient correction of Fabry mice

and patient cells mediated by lentiviral transduction of hematopoietic stem/progenitor

cells. Gene Ther. 14, 256-265 (2007).

17. B. M. Leung and M. V. Sefton, A modular tissue engineering construct containing

smooth muscle cells and endothelial cells. Ann. Biomed. Eng 35, 2039-2049 (2007).

18. R. Schwarzkopf, M. Shachar, T. Dvir, Y. Dayan, R. Holbova, J. Leor, and S. Cohen,

Autospecies and post-myocardial infarction sera enhance the viability, proliferation, and

maturation of 3D cardiac cell culture

1. Tissue Eng 12, 3467-3475 (2006).

Page 89: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

77

19. M. Scharpfenecker, U. Fiedler, Y. Reiss, and H. G. Augustin, The Tie-2 ligand

angiopoietin-2 destabilizes quiescent endothelium through an internal autocrine loop

mechanism. J. Cell Sci. 118, 771-780 (2005).

20. H. Kobayashi, T. Shimizu, M. Yamato, K. Tono, H. Masuda, T. Asahara, H. Kasanuki,

and T. Okano, Fibroblast sheets co-cultured with endothelial progenitor cells improve

cardiac function of infarcted hearts. J. Artif. Organs 11, 141-147 (2008).

21. R. K. Iyer, J. Chui, and M. Radisic, Spatiotemporal tracking of cells in tissue-engineered

cardiac organoids. J. Tissue Eng Regen. Med. 3, 196-207 (2009).

22. A. Lesman, M. Habib, O. Caspi, A. Gepstein, G. Arbel, S. Levenberg, and L. Gepstein,

Transplantation of a tissue-engineered human vascularized cardiac muscle. Tissue Eng

Part A 16, 115-125 (2010).

23. R. K. Iyer, L. L. Chiu, and M. Radisic, Microfabricated poly(ethylene glycol) templates

enable rapid screening of triculture conditions for cardiac tissue engineering. J. Biomed.

Mater. Res. A 89, 616-631 (2009).

24. M. D. Chamberlain, R. Gupta, and M. V. Sefton, Chimeric vessel tissue engineering

driven by endothelialized modules in immunosuppressed Sprague-Dawley rats. Tissue

Eng Part A (2010).

25. R. K. Iyer, M. Radisic, C. Cannizzaro, and G. Vunjak-Novakovic, Synthetic oxygen

carriers in cardiac tissue engineering. Artif. Cells Blood Substit. Immobil. Biotechnol. 35,

135-148 (2007).

26. M. Radisic, A. Marsano, R. Maidhof, Y. Wang, and G. Vunjak-Novakovic, Cardiac

tissue engineering using perfusion bioreactor systems. Nat. Protoc. 3, 719-738 (2008).

27. D. Kaigler, Z. Wang, K. Horger, D. J. Mooney, and P. H. Krebsbach, VEGF scaffolds

enhance angiogenesis and bone regeneration in irradiated osseous defects. J. Bone

Miner. Res. 21, 735-744 (2006).

Page 90: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

78

28. L. Chen, Z. He, B. Chen, M. Yang, Y. Zhao, W. Sun, Z. Xiao, J. Zhang, and J. Dai,

Loading of VEGF to the heparin cross-linked demineralized bone matrix improves

vascularization of the scaffold. J. Mater. Sci. Mater. Med. (2009).

29. H. Naito, I. Melnychenko, M. Didie, K. Schneiderbanger, P. Schubert, S. Rosenkranz, T.

Eschenhagen, and W. H. Zimmermann, Optimizing engineered heart tissue for

therapeutic applications as surrogate heart muscle. Circulation 114, I72-I78 (2006).

30. J. M. Kelm, V. Djonov, S. P. Hoerstrup, C. I. Guenter, L. M. Ittner, F. Greve, A.

Hierlemann, C. D. Sanchez-Bustamante, J. C. Perriard, E. Ehler, and M. Fussenegger,

Tissue-transplant fusion and vascularization of myocardial microtissues and

macrotissues implanted into chicken embryos and rats. Tissue Eng 12, 2541-2553

(2006).

31. M. J. van Luyn, R. A. Tio, X. J. van Seijen, J. A. Plantinga, L. F. de Leij, M. J.

DeJongste, and P. B. van Wachem, Cardiac tissue engineering: characteristics of in

unison contracting two- and three-dimensional neonatal rat ventricle cell (co)-cultures.

Biomaterials 23, 4793-4801 (2002).

Page 91: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

79

Chapter 4: Fate of modular cardiac constructs in vivo

4.1 Abstract

Cardiac tissue engineering holds the promise of restoring cardiac function by replacing

scar tissue with functional cells. The modular tissue engineering approach has been

demonstrated in a previous study to be a useful method for generating functional cardiac tissue

in vitro. However the effects of host immune response and remodeling on modular cardiac

tissues remain unclear. In this study, the fates of modular cardiac tissues in vivo were examined

in two implantation strategies based on a syngeneic animal model. CM-only or co-culture (CM

and EC) modules were either injected into the peri-infarct zone of the heart, or fabricated into

patch form and implanted over a right ventricular free wall defect. After 2 weeks (3 weeks for

patch implants), donor EC developed into blood vessels-like structures, which appeared

functional and connected with host vasculature. However, no donor CM was found within the

implant sites. Also, host immune cells including macrophages and T-cells infiltrated extensively

into implantation site in both models. To lessen host immune response, MatrigelTM was omitted

from matrix and modules were rinsed with serum-free medium prior to implantation. The

removal of xenoprotein attenuated host immune cell infiltration, while donor EC appeared

unaffected and formed functional blood vessels. Co-culture implants had a higher overall vessel

density compared to CM-only implants, but only in the absence of MatrigelTM. Most

importantly, donor CM matured into striated muscle-like structures throughout MatrigelTM-free

implants. In this study we have demonstrated that modular cardiac tissues can be implanted into

the myocardium in multiple configurations and are able to develop into native-like structures in

vivo. The results also suggest that host immune response play a crucial role in the survival and

maturation of implanted modular cardiac tissue.

Page 92: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

80

4.2 Introduction

Myocardial infarction (MI) is caused by the interruption of blood supply to the heart and

can lead to massive cell death. The extracellular matrix and cell debris that are left behind

undergo extensive remodeling mediated by immune cells and resident myofibroblasts. The

inflammatory region eventually stabilizes into collagen-rich, acellular and non-contractile scar

tissue. The scar tissue reduces the overall contractile force of the heart, and at the same time

their inelastic nature increases overall cardiac load. Therefore there is a need to develop

therapeutic strategies that aim to replace scar tissue with functional tissue and ultimately restore

cardiac function.

Current research focuses on two major strategies to achieve this goal. The first is by

injecting cells, either differentiated myocytes1-4 or undifferentiated cardiac progenitor cells

(embryonic5,6 or mesenchymal7-9 stem cells) into or around the infracted region in an attempt to

repopulate the site with functional cells. The second strategy aims to replace the scar tissue with

engineered cardiac muscles consisting of myocytes (CM10-12 or skeletal myoblast13,14) and other

supporting cells types, including EC and fibroblast15,16. One of the obstacles in these strategies is

low cell survival caused by a combination of poor cell engraftment and lack of functional

vasculature within the implant sites. A novel vascularization technique was demonstrated by our

laboratory when EC seeded collagen modules formed functional blood vessels in a rat omental

pouch model after 21 days17. It is proposed that a similar method can create a scalable cardiac

construct with uniformly distributed EC that may accelerate graft vascularization in vivo.

The goal of this study is to devise methods to implant the modular cardiac construct and

to assess their fates in vivo using a syngeneic rat model. Two implantation methods were

investigated. In the first method, macroporous modular sheets containing CM and EC were

sandwiched between two pieces of commercially available porous gelatin foam (GelFoam) to

create an implantable patch. This patch was implanted over a transmural defect on the right

ventricular free wall of a healthy Lewis rat heart for 2 weeks. Using this model we were able to

elucidate the biological interaction, such as ECM remodeling, cell maturation and tissue

integration between implanted cardiac modular construct and host tissue. The second method

investigated the delivery of modules into the peri-infarct zone via intracardial injection.

Modules were injected (CM-only or CM/EC co-culture) into the peri-infarct zone of an acute MI

in a Lewis rat. Using this approach we expected to see better cell engraftment and improved

Page 93: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

81

viability compared to matrix-free cell injection therapy. We also expected that CM-embedded

modules implanted into the rat heart would develop more native-like morphology compared to

CM-modules cultured in vitro.

4.3 Materials and methods

4.3.1 Isolation of rat primary CM and EC

Primary CM and EC were isolated from green fluorescent protein (GFP) transgenic

neonatal Lewis rats (Lewis-Tg [EGFP] F455) obtained from the Rat Resources and Research

Center (RRRC, University of Missouri, Columbia, MO). The aorta and atrium were trimmed

away and the remaining tissues were cut into small pieces. The tissues were incubated in bovine

trypsin (0.6mg/mL in HBSS) overnight at 4oC, followed by serial digestion in collagenase type

II (1 mg/mL) at 37oC. CM-rich cell suspensions were pre-plated onto TCPS flasks for 1 hour to

enrich the CM content. The CM enriched cell mixture was loaded onto a Cytospin® slide and

stained for troponin I and prolyl-4-hydroxylase (see below) to identify CM and fibroblasts

respectively: the mixture consisted of 50% cardiomyocytes and 40% fibroblasts. The remaining

10% was presumed to consist of other cardiac cell types, including EC.

Primary rat cardiac EC were isolated from the cells attached on to the TCPS flasks in the

previous step. The CM-rich cell suspensions were replaced with native EC-medium (MCDM-

131 with 10% FBS). After 2 days, attached cells were trypinized and the EC were labeled with

mouse anti-rat CD31 FITC conjugated antibody followed by anti-FITC magnetic beads

(Mylteny Biotech), and labeled cells were separated using a magnetic cell sorting column.

Isolated EC were maintained with native EC medium in fibronectin coated flasks. EC phenotype

was confirmed by Dil-Ac-LDL uptake assay (10µg/mL Dil-Ac-LDL, 4 hours at 37oC).

4.3.2 Modular cardiac patch model

Modular cardiac patch was prepared by placing CM-embedded modular sheets within a

GelFoam pocket. CM embedded modular sheets were fabricated as described in Chapter 3.3.3.

The sheets were stimulated continuously for 5 days (1 Hz, 5V, 2ms, biphasic square pulse). At

Page 94: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

82

the end of day 5, three modular sheets were placed on a piece of GelFoam, measuring 2mm

thick, 6mm wide and 12mm long. The GelFoam was then folded in half length-wise and sutured

at the open corners to create an implantable patch. For CM/EC co-culture patches, CM modular

sheets were seeded with EC (passage 3-5) at the end of the five-day stimulation period, and the

sheets were culture for 1 day in co-culture medium before being fabricated into patches similar

to CM-only sheets.

The modular patch was implanted over a transmural defect on the right ventricular free

wall using methods described the Li group18. Wildtype Lewis rats (12-20 weeks old) were

sedated, intubated and ventilated (Harvard Apparatus, Holliston, MA, USA), and maintained

with 1.5-2.5% isoflurane. The heart was visualized through a median sternotomy. A 5-6 mm

diameter section of the right ventricular (RV) free wall was isolated with a purse-string suture

(with 7-0 polypropylene monofilament suture) to create a “neck” of the bulging RV free wall, as

described before18-20. A 16-guage angiocatheter was passed over the stitch to create a tourniquet

to prevent bleeding when a full thickness circular segment of the RV free wall was removed.

Patches containing modular sheets were laid on top of the resected ventricular wall and then

sutured to the margin of the defect with a purse-string stitch with 7-0 polypropylene

monofilament stitch using an over-and-over running suture technique to close the defect in the

RV free wall. Then the tourniquet was released and the purse-string stitch was removed. The

chest wall was closed with 5-0 silk suture. Patches were explanted after 14 days and fixed in 4%

paraforamldehyde for histological analysis.

4.3.3 Peri-infarct module injection model

Small diameter CM-only modules and CM/EC co-culture modules were cast in

polyethylene tubes with an inside diameter of 540µm and cut to 1mm in length. The mean post

contraction diameter was approximately 220µm. Other than the difference in diameter, these

modules were fabricated in the same way as the ones used for making the modular patch. CM-

only modules were maintained in native CM-medium until implantation at day 7. Co-culture

modules were made by seeding EC (passage 3-5) onto CM embedded modules at day 3 post-

fabrication and kept in co-culture medium from day 3 to 7 in culture before implantation.

MatrigelTM-free modules were made with type I bovine collagen only. Prior to implantation,

they were rinsed and incubated for 1 hour in defined serum-free medium as describe by Kessler-

Page 95: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

83

Icekson et al.21. Briefly, the medium was composed of 1:1 mixture of Ham’s F12 and DMEM

supplemented with 10 mM HEPES at pH 7.3, additional 1mg/mL glucose, 0.1 µM

hydrocortisone, 25 µg/mL insulin, 25 µg/mL transferrin, 1 mg/mL fetuin (all from Sigma).

An acute MI rat model was created by ligating the left descending coronary artery of

wild type Lewis rats (12-20 weeks old) for 7 days prior to modules implantation. Small diameter

modules were injected into the peri-infarct zone using a 28G needle (3 sites per animal, equal

distance apart). Approximately 50 modules were injected per site for a total of 150 modules per

animal. Animals were sacrificed at 21 days post injection and the hearts were fixed in 4%

paraformaldehyde for histological analysis.

4.3.4 Immunostaining and Histological Analysis

Modules cultured in vitro for 7 days were fixed with 4% paraformaldehyde and

permeabilized using 0.2% Triton-X100 solution. Samples were then triple stained for troponin I,

F-actin and cell nucleus. Embedded CM were identified using rabbit anti-rat troponin I

(Chemicon) antibody followed by Alexa FluorTM 488 goat anti-rabbit secondary antibody

(Molecular Probes). Alexa FluorTM 568 phalloidin (Molecular Probes) and Hoechst (Sigma)

were used to stain for actin cytoskeleton and cell nucleus, respectively. Samples were visualized

using a Zeiss LSM510 confocal microscope.

For in vivo analysis, animals were sacrificed at 2 week (patch model) or 3 week (module

injection model) after implantation. Explanted tissues were fixed in 4% paraformaldehyde for

48 hours at 4oC, followed by incubation in 30% sucrose solution for 24 hours. Tissue samples

were rinsed in 70% ethanol before being embedded in paraffin. Tissue sections were stained

with H&E and Masson’s trichrome for tissue morphology assessment. Macrophages and T-cells

were stained with monoclonal mouse anti-rat CD68 (Serotec) and mouse anti rat T cell receptor

(BD Pharagen) antibodies respectively. Implanted cells were tracked by staining tissue sections

with polyclonal rabbit anti-GFP antibody (Abcam), or double staining with GFP and CD31 (or

myosin heavy chain, MHC). For MHC staining tissue section were sequentially stained with

polyclonal rabbit anti-GFP antibody (Abcam) visualized with DAB, followed by monoclonal

mouse anti-rat MHC (α- and β- isoforms, Abcam) antibody visualized with Vector Red. Donor

derived blood vessels were detected by sequentially staining tissue sections with polyclonal goat

anti-rat CD31 antibody (Santa Cruz) visualized with DAB, followed by polyclonal rabbit anti-

Page 96: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

84

GFP antibody (Abcam) visualized with Vector Red. Vessel density for each sample was

determined by averaging the number of target structures in three hot spots under 100x

magnification.

4.3.5 Statistical analysis

Statistical analysis including calculations of standard error of means and mean

comparisons (t-test or one-way ANOVA) were performed using Prism (Version 5.0, Graphpad

Software, La Jolla, CA)

Page 97: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

85

4.4 Results

4.4.1 Characterization of CM embedded modules and rat heart EC

We examined the morphology and viability of CM embedded small diameter modules

after 7 days in culture to establish baseline conditions prior to implantation. Cells on the

perimeter appeared elongated in collagen-only modules as well as in collagen+MatrigelTM

module. However, troponin I positive cells near the core appear rounded (Figure 4-1) compared

to cells on the perimeter.

Primary cardiac microvascular EC from transgenic GFP+ Lewis rats were isolated and

concentrated, using CD31 antibody conjugated magnetic beads after CM harvest. To verify their

phenotype, EC were incubated with Dil-Ac-LDL and visualized by confocal microscopy (Figure

4-2). Over 90% of cells are EC, as indicated by their uptake of Dil-Ac-LDL.

We were interested in determining if rat EC adhesion and junction morphology were

affected during the module injection procedure. To answer this question, small diameter

modules embedded with CM were coated with non-GFP rat aortic EC using similar methods as

before22. At 7 days post seeding, CM/EC modules were loaded into a syringe then ejected

through a 28G disposable needle, similar to the ones used for in vivo injection. Immediately

after injection the modules were fixed and stained for VE-cadherin (Figure 4-3A). Confocal

microscopy showed no significant loss of EC in modules which passed through the needle when

compared with modules that did not pass through the needle (Figure 4-3B). However, EC

junctions did appear to be disturbed.

Page 98: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

86

Figure 4-1: Immunofluorescence staining of CM embedded small diameter modules

supplemented with MatrigelTM after 7 days in static culture containing 10% BS (green - troponin

I, blue - cell nucleus, red - F-actin). Cells on the perimeter appear elongated in both collagen-

Page 99: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

87

only (A,B,C) and collagen+MatrigelTM (D,E,F) modules. In both groups, cell density decreases

toward the core of modules.

Figure 4-2: Immunoflorescence micrograph of rat cardiac EC isolated from transgenic GFP+

Lewis rats. EC were isolated using CD31 conjugated magnetic beads. After 2 passages, EC were

seeded at 50% confluence onto fibronectin coated glass chambered slides and stained with Dil-

Ac-LDL (10µg/mL, 6 hours). EC stained with Dil-Ac-LDL (red) maintained GFP expression

(green).

Page 100: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

88

Figure 4-3: EC coated modules before and after injection through a 28G needle. Matrigel

supplemented CM modules made with small diameter tubing (I.D. = 500µm) were coated with

RAEC and static cultured for 7 days. Modules were loaded into a 1 mL syringe and ejected

through a diposible ½ inch, 28G needle. Modules were fixed and stained immediately. VE-

cadherin staining (green) show no significant cell loss compared to modules before injection,

however, EC junctions appeared more disorganized (100x magnifications).

4.4.2 Modular patch and injection implants

In this study we explored two different implantation strategies to deliver modular cardiac

constructs. In the first strategy an implantable version of the modular sheet was fabricated by

stacking modular sheets between two pieces of collagen foam. Three modular sheets, containing

collagen, MatrigelTM, and cells harvested from transgenic GFP+ Lewis rats, were placed

between collagen foams and the corners were sutured to form a pouch (Figure 4-4A). The pouch

was implanted over the right ventricular free wall of wildtype syngeneic Lewis rats. At 14 days

the right ventricular wall was excised and sectioned for histological analysis.

In the second implanation model, small diameter CM-only and CM/EC co-culture

modules containing MatrigelTM were injected through a 28G needle into the peri-infarct zone of

a 7-days old acute left ventricular infarct. Modules were seen around the injection site, and they

remained visible when explanted after 3 weeks (Figure 4-4B).

Page 101: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

89

Patch and module injection implants were retrieved after 2 week and 3 week,

respectively. In both models, implanted modules were extensively remodeled by host cells, and

in most cases, cannot be identified by histological staining (Figure 4-5). Moreover, there were

significant amount of collagen deposit in the implantation sites by host tissue. Histological

sections also indicate thinning of the heart wall caused by the remodeling of the infarct in the

injection model. (Figure 4-6).

In both implantation models GFP+ cells were found though out the implant sites (Figure

4-7). The presence of EC in the co-culture implant did not lead to a substantial change in GFP+

cell density. Cells that were GFP+/CD31+ formed blood vessel-like structures within the co-

cultured patch (Figure 4-8E and F) and injection (Figure 4-9E and F) implants. In some cases,

blood vessel-like structures were lined with a combination of GFP+/CD31+ and GFP-/CD31+

cells (Figure 4-8F and 8F). Furthermore, red blood cells were found inside the lumen of these

vessels, which suggest that these vessels may have connected with host vasculature and were

functional. Similar structures were also found in CM-only implants in both models. However,

these vessels were lined with GFP-/CD31+ cells only, suggesting that the EC were host-derived

(Figure 4-8B,C and 4-9B,C).

Lastly we stained explanted tissues for GFP and myosin heavy chains (MHC, α- and β-

isoforms) to evaluate the fate of implanted CM. MHC+ structure was found in neither the CM-

only or co-culture patch implants (Figure 4-10A and D). In module injection implants, GFP+/

MHC+ cells in muscle like bundles were found in CM-only implants (Figure 4-10B and C), but

not in co-culture implants (Figure 4-10E and F).

Page 102: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

90

Figure 4-4: Schematic diagrams and tissue morphology at explant for modular patch implant

module injection implant. (A) Patch implants were created by placing 3 modular sheets between

two pieces of GelFoam and were sutured over a transmural defect on the right ventricular free

wall of a wildtype Lewis rat. (B) In a different model, individual small diameter modules were

injected into the peri-infract zone (3 sites, 50 modules/site) of an 7 day old MI (white dash line)

in a wildtype Lewis rat using a 28G needle. Modules (white arrow) can be found around the

border zone (yelow dotted line) outside of the infarcted area (white dotted line) when explanted

after 3 weeks.

Page 103: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

91

Figure 4-5: Masson’s trichrome stain of patch implants after 2 weeks. MatrigelTM supplemented

collagen modules in all groups underwent extensive host remodeling. Large deposits of collagen

and granulation tissues were found within the GelFoam matrix in CM-only (A and B) and co-

culture (C and D) modular patch implants. In some cases, implanted modules could be identified

morphologically (D, white dotted line).

Page 104: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

92

Figure 4-6: Masson’s trichrome stain of module injection implants after 3 weeks. MatrigelTM

supplemented collagen modules injected into the peri-infarct region underwent extensive host

remodeling.

Page 105: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

93

Figure 4-7: Immunohistochemical staining of explants revealed the presence of GFP+ donor

cells scattered within the implant site in patch implants (A and C) and module injection implants

(B and D). The expression of GFP suggests implanted cells remained viable. The presence of

EC in co-culture implants did not translate into higher GFP+ cell density (slides shown at 100x

magnification).

Page 106: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

94

Figure 4-8: Blood vessel-like structures can be found in both CM-only (A,B and C) and co-

culture (D, E and F) modular patch implants. The presence of red blood cells suggests vessels

are functional. Blood vessels were evenly distributed within the modular cardiac construct. High

magnification (400x) revealed blood vessels in co-culture patches are lined entirely (E) or

partially (F) with GFP+/CD31+ donor derived EC (pink and brown). In CM-only modular patch

implants, most cells lining the vessels are CD31+ but GFP-, suggesting that they are derived

from host EC.

Page 107: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

95

Figure 4-9: Blood vessel-like structures can be found in both CM-only (A,B and C) and co-

culture (D, E and F) injection implants. The presence of red blood cells suggests vessels are

functional. Higher magnification (400x) revealed blood vessels were lined entirely (E) or

partially (F) with GFP+/CD31+ donor derived EC (pink and brown) in co-culture injection

implants. In CM-only implants (B and C), most vessels are lined with cells that are CD31+ but

GFP-, suggesting they are derived from host EC.

Page 108: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

96

Figure 4-10: Double staining for GFP (brown) and MHC (pink) revealed donor derived CM

developed into striated muscle like structure in CM-only injection implant (B, 100x and C,

400x). These double stained, GFP+/ MHC+ structures were not found in other groups (co-culture

patch – D, CM-only patch – A, and co-culture module injection – E,F)

Page 109: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

97

4.4.3 Host immune response and the removal of MatrigelTM

Host responses towards implanted constructs can significantly affect construct function

and survival. In this study we examined these responses by staining for macrophages and T-cell.

The presence of MatrigelTM in the patch implants and the module injection implants elicited a

strong macrophage response (Figure 4-11A, B, D and E) as well as T-cell infiltration (Figure 4-

12A, B, D and E). We attempted to attenuate these immune responses in the module injection

implant model by removing MatrigelTM from the matrix and rinsing the modules in serum-free

medium prior to implantation. After 3 weeks these implants were removed and examined by

histology. There were substantially less macrophage and T-cell infiltration into the implants in

the absence of MatrigelTM (Figure 4-11C, F and 4-12C, F). Blood vessel like structures lined

with GFP+/CD31+ cells were found through out CM/EC co-culture implants. In contrast, similar

structures found in CM-only implants were lined with GFP-/CD31+ cells (Figure 4-13).

When quantified, patches containing CM-only modules and co-culture modules showed

no difference in total vessel density (Figure 4-15A). But when the vessel are grouped based on

cell phenotype, we found an increased number of vessels lined with GFP+/CD31+ cells in the co-

culture patch implants compared to CM-only patches (Figure 4-15B). Also, co-culture implants

without MatrigelTM have significantly higher vessel density counts compared to co-culture

implants with MatrigelTM and CM-only MatrigelTM-free implants (Figure 4-15A). However, the

removal of MatrigelTM did not alter the proportion of GFP+ vessels found in the implants

compared to those with MatrigelTM (Figure 4-15B).

The removal of MatrigelTM also affected muscle bundle formation. In both CM-only and

co-culture implants, we found an abundance of muscle-like structures throughout the implants.

Unlike implants with MatrigelTM, many of these bundles contained GFP+/MHC+ cells (Figure 4-

14).

Page 110: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

98

Figure 4-11: Immunohistochemical staining of CD68 revealed large number of macrophage

infiltration in all implantation models with MatrigelTM (A,B,D and E, 100x magnification).

MatrigelTM-free CM-only (C) and co-culture (F) injection implants lead to a significant

reduction in macrophage infiltration after 3 weeks, as shown by the reduction of CD68+ cells.

Page 111: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

99

Figure 4-12: Immunohistochemical staining of T cell receptor (TCR) revealed large number of

T-cell infiltration in all implantation models with MatrigelTM (A,B,D and E, 100x

magnification). It was not known whether these T-cells were activated or not. MatrigelTM-free

CM-only (C) and co-culture (F) injection implants lead to a significant reduction in T-cell

infiltration after 3 weeks, as shown by the reduction of TCR+ cells.

Page 112: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

100

Figure 4-13: Blood vessel-like structures were found in MatrigelTM-free CM-only (A and B)

and co-culture (C and D) injection implants after 3 weeks. Under higher magnification (400x),

blood vessels comprise partially of GFP+/CD31+ donor derived EC (pink and brown) were found

in co-culture injection implants (D). In CM-only patches (B), vessels are lined with GFP-/CD31+

cells (brown), suggesting the vessels arise from host derived EC. The presence of red blood cells

in the lumens suggests the vessels connected to host vasculature and are functional.

Page 113: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

101

Figure 4-14: Donor CM in MatrigelTM-free CM-only (A and B) and co-culture (C and D)

injection implants formed striated muscle-like structures after 3 weeks. Under higher

magnification (400x) striated muscle bundles were found. These structures contain both host

derived CM (GFP-/MHC+, pink) as well as donor derived CM (GFP+/ MHC+, pink and brown).

Page 114: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

102

Figure 4-15: Total vessel density counts show no significant difference in vessel densities

between CM-only constructs and their co-culture counterparts (A). In MatrigelTM-free injection

implants (CM-Injection-MF and CO-Injection-MF), however, the presence of EC significantly

increased vessel density compared to CM-only implants and co-culture injection implants with

MatrigelTM (*, p<0.05, n=3). In all cases, donor cells participated in vessel formation, as shown

by the increased proportion of GFP+ and mixed vessel when counted based on their phenotypes

(GFP+ EC only, mixed EC, or GFP- EC only).

Page 115: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

103

4.5 Discussion

4.5.1 Implantation strategies

In previous studies we have demonstrated that modules consisting of MatrigelTM

supplemented collagen were able to maintain the viabilities and functionalities of embedded CM

and surface coated EC. We have also shown that the modules could be used to form

macroporous sheets in vitro22. In this study our goal was to create implantable constructs using

the modular approach and determine their fates in vivo. Our first approach was to assemble

module sheets into an implantable patch. However, these sheets did not posses the necessary

mechanical strength to be surgically handled as a stand alone patch; therefore we had to devise a

way to support the module sheets. The method described in this study involved sandwiching

module sheets between two pieces of GelFoam, a commercially available sponge-like substrate

made of gelatin. The GelFoam layers provided mechanical stability to the patch construct so that

it could be sutured onto the right ventricular free wall defect. Histological analysis revealed that

a significant number of host cells including macrophage, T-cells and fibroblast infiltrated into

the GelFoam after 2 weeks. These cells resorbed and remodeled much of the GelFoam similar to

what was observed in other studies involving collagen or gelatin foam scaffolds19,23. We found

most of the GFP+ cells clustered in module-shaped areas within a collagen rich deposit, and

almost none of them were found near the host-implant interface, where the tissue type is

predominantly host derived. The presence of EC on co-culture modules did not seem to have a

noticeable effect on the gross tissue remodeling outcome.

Aside from using the module sheets to create constructs to cover a full thickness defect,

we also explored the possibility of deploying modules as individual functionalized microtissues

to repopulate damaged cardiac tissue. To test this approach CM-only or co-culture modules

were injected into the peri-infarct region of an acute infarct in the left ventricle. The peri-infarct

zone was chosen due to its location between the infarct zone and healthy myocardium. At this

location, the modules will be more likely to survive, being adjacent to oxygenated tissue, while

remaining close to the infarct zone to exert its therapeutic effects. Histological analysis after 3

weeks showed heart wall thinning and decellularization within the infarct zone. Modules were

found in the peri-infarct zone with most of their collagen matrix still intact. When compared to

the patch implant model, the injection model has a much smaller host-tissue transition zone, and

Page 116: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

104

the modules are separated from the host tissue by only a thin layer of collagen. In the absence of

GelFoam, the host tissue remained organized and few myofibroblasts infiltrated into implants.

Like the patch implants a large amount of macrophage and T-cells infiltrated into the implants

sites, suggesting that the use of GelFoam in the patch used may not be sole factor for triggering

the larger than expected host immune response.

These two implantation strategies were intended to highlight the potential applications of

the modular cardiac construct. The patch implant was initially designed to cover full thickness

defect, and as such it should possess the necessary mechanical strength. In its current form the

module sheet construct cannot easily be handled or sutured into native myocardium. The

addition of a GelFoam layers solved this problem, but at the same time it may act as a barrier for

proper implant-host integration by separating the modular sheets from the wound edge of the

RVOT defect. Moreover, the addition of GelFoam decreased the overall cell-to-scaffold ratio

when compared to constructs consisting of modules alone, thus potentially reducing the

theoretical force that could be generated by such implant. Therefore, more work is still needed

to fabricate the modules into a clinically useful cardiac patch implant.

Alternatively we deployed loose CM-embedded modules as pre-fabricated tissue unit

into the peri-infarct zone in an MI; an area that has undergone an ischemic event and

experienced heavy cell loss. The rationale here is similar to that of cell therapies based on

intramyocardial cell injection, where the goal is to rapidly repopulate functional cell types

including CM and EC. The potential advantages of injecting modules over cell suspension may

be better cell retention and cell survival. Indeed, the modules were physically lodged within the

injection site and we saw no modules escaping through the needle puncture hole after implant.

Histology sections confirmed the presence of module and viable cells after 3 week. On the other

hand, when compared with scaffold free cell injection methods, the presence of collagen may

impede with the integration and assembly of CM into muscle bundles. Whether the modular

injection method of cell delivery into the myocardium is superior to conventional cell injection

remains to be determined by functional assays in future studies.

Page 117: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

105

4.5.2 Fates of implanted cells and host immune response

The ultimate goal of cardiac tissue engineering is to restore lost function to the heart.

Whether or not this goal is achieved depends on the survival and function of implanted cells as

well as their interactions with host tissue. Specifically, the new tissue must contain contractile

CM and the vasculature to support them. The fates of implanted cells were examined by

histological analysis.

Previous studies have demonstrated that allogeneic EC coated on collagen modules were

capable of forming functional blood vessels in an immunosuppressed animal after 3 weeks17. In

this study we saw similar vessel-like structures within the modular patch implants and injected

modules implants. It is likely that these vessels have anastamosed with host vasculature and are

functional, as erythrocytes were found within their lumens. In both cases, we found vessels lined

with GFP+/CD31+ cell when co-culture modules were implanted, which suggests that these

vessels were derived from implanted EC. Also, vessels consisted of host derived EC alone or a

mixture of host and implanted EC were found within the vicinity of implants. These

observations suggest that the implanted EC not only form vascular structures, but may also be

involved in creating and maintaining an angiogenic gradient to attract host EC migration and

play a role in stabilizing new blood vessels formation around the implant site10. The factors

responsible for the observed angiogenic events remain to be determined in future studies.

We hypothesized that the presence of EC in co-culture implants would lead to an

increase in overall vessel density. Indeed this was the case in the modular injection implant

model, but only in the absence of MatrigelTM. It is possible that host immune response against

xenoprotein may have masked the difference in vessel density between CM-only and co-culture

implants. Alternatively, the presence of growth factors in implants containing MatrigelTM may

have mobilized host EC and overwhelmed the angiogenic effect of donor EC, resulting in

similar overall vessel density in both implants.

The presence of xenoprotein also had an effect on the overall vessel density amongst

similar implants. MatrigelTM-free co-culture implants had a higher overall vessel density

compared to co-culture implants with MatrigelTM. This suggests that the host immune response

against implants with MatrigelTM may have diminished the angiogenic potential of donor EC.

The main motivation in building a functional vasculature in engineered cardiac tissue is

to support the metabolic needs of CM, which are essential for their development and

Page 118: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

106

organization into differentiated muscles, as several studies have highlighted this point15,16,24. In

this study, we looked at the expression of MHC as the primary marker of tissue development.

With the exception of CM-only injected module, few implanted CM survived. However, those

that did survive developed into striated muscle like structures in vivo in modules containing

MatrigelTM. This suggests that the maintenance of CM survival shortly after implantation may

be the key for successful tissue formation.

The low CM survival rate was accompanied by a strong macrophage and T-cell

infiltration. It is important to recognize that the recruitment of macrophage and the associated

inflammatory response are crucial for the proper wound healing and revascularization process.

Not only do macrophages remove cell debris from the trauma site, certain subpopulation of

macrophages, namely the M2 sub-type, are known to secrete VEGF and other factors to promote

angiogenesis25. However, an overactive inflammatory response brought on by non-self proteins

may impair cell survival. For this reason, we have chosen to perform all implantation studies

using syngeneic animals in order to minimize the immune response caused by major

histocompatibility complex mismatch between donor and host. Even though all cells were

harvested from syngeneic donors, the MatrigelTM and bovine serum used during culture may

have aggravated the inflammatory response and triggered the activation of T-cells, leading to a

cytotoxic environment that contributed to CM death. Other studies have shown that the presence

of xenogeneic protein in MatrigelTM and bovine serum used during culture can provoke a strong

host immune response13,26. In particular, a study by Zimmermann et al. showed that the

immunogenicity of MatrigelTM elicited an immune response that can significantly reduce

implanted CM survival12. It is likely that the implanted CM in MatrigelTM+collagen modules

suffered the same fate. Not surprisingly, the removal of MatrigelTM and pre-implantation rinse

of modules in serum-free medium substantially reduced the amount of macrophage and T-cell

infiltration. Along with the reduction in host immune response was an increase in CM survival

and the presence of GFP+/MHC+ striated muscle structure in both CM-only and co-culture

injection implants. However, it appeared that the presence of EC in co-culture injection implants

did not lead to a significant difference in CM survival after 3 weeks. Perhaps the benefit of EC

will be more pronounced in later time points.

Beside improved CM survival, the removal of xenoproteins also improved blood vessel

density. Module injection with collagen-only, co-culture modules had a higher vessel density

compared to the CM-only group. In fact, the difference in vessel density was only present in the

collagen-only implants. It is possible that host immune response triggered by MatrigelTM and

Page 119: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

107

bovine serum have also lead to the death of implanted EC, similar to the fate of implanted CM.

These results highlight the issue of using xenomaterial as part of the cardiac scaffold for clinical

applications. Development in synthetic scaffolds and hydrogel based on autologous ECM may

mitigate this problem27.

4.6 Conclusion

In this study we have demonstrated that modular cardiac tissues could be

implanted successfully in its basic form or as a patch implant in a syngeneic animal model. In

both implantation strategies, donor EC formed blood vessels-like structures and appeared to

have anastamosed with host vasculature. In co-culture implants, these vessels were lined with a

mixture of donor and host derived EC. In contrast, vessels found in CM-only implants only

contain host derived EC. The presence of MatrigelTM and bovine serum in CM-only and co-

culture implants triggered significant host immune and inflammatory responses which may be

responsible for the loss of implanted CM in both the patch and injection model. The subsequent

removal of foreign protein substantially reduced macrophage and T-cell infiltration into the

implants. Co-culture implants had a higher overall vessel density compared to CM-only

implants, but only in the absence of MatrigelTM. It also led to an increase in striated muscles

bundles containing donor CM. These results will serve as the basis for future optimization

studies toward the realization of the modular cardiac construct as clinically relevant method to

replace damaged cardiac tissue.

Page 120: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

108

Reference List

1. W. Sherman, K. L. He, G. H. Yi, J. Wang, J. Harvey, M. J. Lee, H. Haimes, P. Lee, E.

Miranda, S. Kanwal, and D. Burkhoff, Myoblast transfer in ischemic heart failure:

effects on rhythm stability. Cell Transplant. 18, 333-341 (2009).

2. T. Patila, T. Ikonen, E. Kankuri, A. Uutela, J. Lommi, L. Krogerus, P. Salmenpera, J.

Bizik, K. Lauerma, and A. Harjula, Improved diastolic function after myoblast

transplantation in a model of ischemia-infarction. Scand. Cardiovasc. J. 43, 100-109

(2009).

3. P. Farahmand, T. Y. Lai, R. D. Weisel, S. Fazel, T. Yau, P. Menasche, and R. K. Li,

Skeletal myoblasts preserve remote matrix architecture and global function when

implanted early or late after coronary ligation into infarcted or remote myocardium.

Circulation 118, S130-S137 (2008).

4. P. Farahmand, T. Y. Lai, R. D. Weisel, S. Fazel, T. Yau, P. Menasche, and R. K. Li,

Skeletal myoblasts preserve remote matrix architecture and global function when

implanted early or late after coronary ligation into infarcted or remote myocardium.

Circulation 118, S130-S137 (2008).

5. J. Nussbaum, E. Minami, M. A. Laflamme, J. A. Virag, C. B. Ware, A. Masino, V.

Muskheli, L. Pabon, H. Reinecke, and C. E. Murry, Transplantation of undifferentiated

murine embryonic stem cells in the heart: teratoma formation and immune response.

FASEB J. 21, 1345-1357 (2007).

6. M. A. Laflamme, J. Gold, C. Xu, M. Hassanipour, E. Rosler, S. Police, V. Muskheli, and

C. E. Murry, Formation of human myocardium in the rat heart from human embryonic

stem cells. Am. J. Pathol. 167, 663-671 (2005).

7. L. C. Amado, A. P. Saliaris, K. H. Schuleri, M. St John, J. S. Xie, S. Cattaneo, D. J.

Durand, T. Fitton, J. Q. Kuang, G. Stewart, S. Lehrke, W. W. Baumgartner, B. J. Martin,

A. W. Heldman, and J. M. Hare, Cardiac repair with intramyocardial injection of

Page 121: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

109

allogeneic mesenchymal stem cells after myocardial infarction. Proc. Natl. Acad. Sci. U.

S. A 102, 11474-11479 (2005).

8. A. B. Mathiasen, M. Haack-Sorensen, and J. Kastrup, Mesenchymal stromal cells for

cardiovascular repair: current status and future challenges. Future. Cardiol. 5, 605-617

(2009).

9. C. H. Yoon, M. Koyanagi, K. Iekushi, F. Seeger, C. Urbich, A. M. Zeiher, and S.

Dimmeler, Mechanism of Improved Cardiac Function After Bone Marrow Mononuclear

Cell Therapy. Role of Cardiovascular Lineage Commitment. Circulation (2010).

10. H. Sekine, T. Shimizu, K. Hobo, S. Sekiya, J. Yang, M. Yamato, H. Kurosawa, E.

Kobayashi, and T. Okano, Endothelial cell coculture within tissue-engineered

cardiomyocyte sheets enhances neovascularization and improves cardiac function of

ischemic hearts. Circulation 118, S145-S152 (2008).

11. W. H. Zimmermann, M. Didie, G. H. Wasmeier, U. Nixdorff, A. Hess, I. Melnychenko,

O. Boy, W. L. Neuhuber, M. Weyand, and T. Eschenhagen, Cardiac grafting of

engineered heart tissue in syngenic rats. Circulation 106, I151-I157 (2002).

12. W. H. Zimmermann, I. Melnychenko, G. Wasmeier, M. Didie, H. Naito, U. Nixdorff, A.

Hess, L. Budinsky, K. Brune, B. Michaelis, S. Dhein, A. Schwoerer, H. Ehmke, and T.

Eschenhagen, Engineered heart tissue grafts improve systolic and diastolic function in

infarcted rat hearts. Nat. Med. 12, 452-458 (2006).

13. M. N. Giraud, E. Ayuni, S. Cook, M. Siepe, T. P. Carrel, and H. T. Tevaearai, Hydrogel-

based engineered skeletal muscle grafts normalize heart function early after myocardial

infarction. Artif. Organs 32, 692-700 (2008).

14. Y. Itabashi, S. Miyoshi, S. Yuasa, J. Fujita, T. Shimizu, T. Okano, K. Fukuda, and S.

Ogawa, Analysis of the electrophysiological properties and arrhythmias in directly

contacted skeletal and cardiac muscle cell sheets. Cardiovasc. Res. 67, 561-570 (2005).

15. A. Lesman, M. Habib, O. Caspi, A. Gepstein, G. Arbel, S. Levenberg, and L. Gepstein,

Transplantation of a tissue-engineered human vascularized cardiac muscle. Tissue Eng

Part A 16, 115-125 (2010).

Page 122: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

110

16. K. R. Stevens, K. L. Kreutziger, S. K. Dupras, F. S. Korte, M. Regnier, V. Muskheli, M.

B. Nourse, K. Bendixen, H. Reinecke, and C. E. Murry, Physiological function and

transplantation of scaffold-free and vascularized human cardiac muscle tissue. Proc.

Natl. Acad. Sci. U. S. A 106, 16568-16573 (2009).

17. M. D. Chamberlain, R. Gupta, and M. V. Sefton, Chimeric vessel tissue engineering

driven by endothelialized modules in immunosuppressed Sprague-Dawley rats. Tissue

Eng Part A (2010).

18. T. Sakai, R. K. Li, R. D. Weisel, D. A. Mickle, E. T. Kim, Z. Q. Jia, and T. M. Yau, The

fate of a tissue-engineered cardiac graft in the right ventricular outflow tract of the rat. J.

Thorac. Cardiovasc. Surg. 121, 932-942 (2001).

19. T. Ozawa, D. A. Mickle, R. D. Weisel, N. Koyama, S. Ozawa, and R. K. Li, Optimal

biomaterial for creation of autologous cardiac grafts. Circulation 106, I176-I182 (2002).

20. T. Ozawa, D. A. Mickle, R. D. Weisel, N. Koyama, H. Wong, S. Ozawa, and R. K. Li,

Histologic changes of nonbiodegradable and biodegradable biomaterials used to repair

right ventricular heart defects in rats. J. Thorac. Cardiovasc. Surg. 124, 1157-1164

(2002).

21. G. Kessler-Icekson, O. Sperling, C. Rotem, and L. Wasserman, Cardiomyocytes cultured

in serum-free medium. Growth and creatine kinase activity. Exp. Cell Res. 155, 113-120

(1984).

22. B. M. Leung and M. V. Sefton, A Modular Approach to Cardiac Tissue Engineering.

Tissue Eng Part A (2010).

23. R. K. Li, Z. Q. Jia, R. D. Weisel, D. A. Mickle, A. Choi, and T. M. Yau, Survival and

function of bioengineered cardiac grafts. Circulation 100, II63-II69 (1999).

24. J. M. Kelm, V. Djonov, S. P. Hoerstrup, C. I. Guenter, L. M. Ittner, F. Greve, A.

Hierlemann, C. D. Sanchez-Bustamante, J. C. Perriard, E. Ehler, and M. Fussenegger,

Tissue-transplant fusion and vascularization of myocardial microtissues and

macrotissues implanted into chicken embryos and rats. Tissue Eng 12, 2541-2553

(2006).

Page 123: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

111

25. A. Sica, P. Larghi, A. Mancino, L. Rubino, C. Porta, M. G. Totaro, M. Rimoldi, S. K.

Biswas, P. Allavena, and A. Mantovani, Macrophage polarization in tumour progression.

Semin. Cancer Biol. 18, 349-355 (2008).

26. Y. Yildirim, H. Naito, M. Didie, B. C. Karikkineth, D. Biermann, T. Eschenhagen, and

W. H. Zimmermann, Development of a biological ventricular assist device: preliminary

data from a small animal model. Circulation 116, I16-I23 (2007).

27. S. B. Seif-Naraghi, M. A. Salvatore, P. J. Schup-Magoffin, D. P. Hu, and K. L.

Christman, Design and characterization of an injectable pericardial matrix gel: a

potentially autologous scaffold for cardiac tissue engineering. Tissue Eng Part A 16,

2017-2027 (2010).

Page 124: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

112

Chapter 5: Future Perspectives and Conclusions

5.1 Conclusions

The overall goal of this project was to apply the modular tissue engineering construct to

create functional cardiac tissues. To begin, we fabricated and characterized a co-culture modular

cardiac tissue in vitro containing neonatal CM and EC. Modules made of type I collagen or

MatrigelTM supplemented collagen were able to support CM growth and function for 14 days in

culture. Modules contracted synchronously in response to external electrical stimulation. The

presence of MatrigelTM increased contraction amplitude and decreased excitation threshold.

Similar functional improvements could be achieved when using medium supplemented with

10% bovine serum instead of 10% fetal bovine serum. Culture conditions for co-culture modules

were determined by fine tuning medium compositions to satisfy the need of both embedded CM

and surface seeded EC so that both cells types maintained their phenotype while in culture.

After 14 days in culture, surface seeded EC covered the entire surface of the module and

expressed VE-cadherin while embedded CM exhibited decreased electrical responsiveness

(excitation threshold, maximum capture rate), but there was no reduction in contraction

amplitude compared to CM-only modules. These studies helped to establish the baseline

conditions for fabricating functional cardiac modules.

The next step was to build on the modular concept and fabricate larger constructs with

an eventual goal of achieving the scale and complexity of native myocardium. We created

macroporous modular sheets by immobilizing co-culture modules in alginate gel in vitro for 10

days, while simultaneously subjecting the sheet to electrical stimulation during culture. Over

this period, adjacent modules fused with one another to form an integrated sheet, while

intermodular spaces were preserved by the alginate gel. When released from the alginate gel,

modular sheets contracted synchronously in response to external electrical stimulation.

Histological analysis revealed many embedded CM expressing troponin I and connexion-43

around the perimeter of modules as well as at intermodular junctions. Morphologically, CM

around the perimeter of modules appeared aligned and elongated. Their resemblance to native

muscle bundles suggested that these structures may be responsible for the generation of the

observed contractile force. Overall, these results demonstrated the feasibility of building higher

Page 125: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

113

order structures (albeit small ones) using the modular approach as well as the structural and

functional integration of modules into a modular sheet.

In the last study, we investigated the fate of modular cardiac tissue in the myocardium.

Two syngeneic implantation models, namely patch and injection models, were used to study the

remodelling of cardiac modules and their interactions with host tissue. Co-culture modules

containing MatrigelTM and collagen were either injected into the peri-infarct zone of the heart or

fabricated into a patch form and implanted over a defect on the right ventricular free wall.

Explanted tissues indicated that donor cells remained viable (2 weeks for patch and 3 weeks for

injection), while host remodelling of collagen matrix was observed. In both models, donor EC

along with other cells, developed into blood vessel-like structures that appeared to connect with

host vasculature. In CM-only implants, blood vessels lined with host derived EC were found

throughout the implant. Therefore, the presence of donor EC in co-culture implants did not lead

to an overall increase in implant vascularity compared to CM-only implants. Interestingly, no

donor CM were found in either model, which suggested that they may have died. Also, host

immune/inflammatory cells such as macrophages and T-cells were found in or near the implant.

To lessen the host response, collagen-only co-culture modules were rinsed in serum-free

medium prior to peri-infarct implantation. Removal of xenoproteins attenuated host immune

response by reducing macrophage and T-cell infiltration into implants. Unlike previous

observations, co-culture implants had a significantly higher vessel density compared to CM-only

implants in the absence of MatrigelTM. In fact, MatrigelTM-free co-culture implants had a higher

overall vessel density compared to co-culture implants with MatrigelTM. Most importantly, in

vivo, donor CM matured into striated, muscle-like structure throughout the implant. This was a

significant improvement over implants with MatrigelTM and the results suggested that early host

response plays a significant role in the survival and maturation of modular cardiac constructs.

The studies presented in this thesis demonstrated the utility of the modular construct as a

means for combining EC and CM in a spatially defined and functional subunit. The results

highlighted the novelty and challenges of building scalable cardiac-tissue equivalence using the

modular approach. Finally, data from in vivo studies showed that cardiac modular tissue

implanted into the myocardium survives and undergoes extensive remodelling and maturation;

events that are mediated by host immune response. These studies have laid the groundwork for

the development of modular cardiac tissue and their use as a therapeutic platform.

Page 126: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

114

5.2 Recommendations

The modular tissue engineering construct was able to solve some of the challenges in

creating large 3-D tissue. However, many obstacles remain to be overcome. Some of these

issues relate specifically to the modular construct, while others are more general and apply to

most tissue engineering strategies. These issues, along with some recommended studies to

address them, are discussed in the following sections.

5.2.1 Improving embedded cell viability

The maintenance of cell viability has been a major challenge in designing engineering

tissue constructs. In many cases, the lack of a vasculature in constructs limits both the size and

complexity of in vitro engineered tissue. The modular approach to engineering tissue constructs

was designed primarily to address this issue by reducing the maximum distance between

embedded cells and the perimeter of the construct to within the theoretical nutrient diffusion

limit of 100-200 µm. Nevertheless, we saw a decline in the density of viable cells towards the

core of modules (diameters ≤ 400 µm). In culture, embedded CM near the core of modules

expressed troponin I, but did not mature into elongated and striated muscle bundles like those

found near the perimeter. This observation may imply that the effective diffusion limit in CM

embedded modules may be much less than expected.

One of the major factors that causes CM death in tissue engineering constructs is

hypoxia. Cells embedded near the module core may suffer from hypoxia due to diffusion

limitation. Core hypoxia can be confirmed by monitoring the expression of hypoxia inducibled

factor 1α (HIF-1α), either by immunohistochemistry or PCR. HIF-1α is a transcription factor

expressed under reduced oxygen tension1. Its expression has been linked to the up regulation of

genes including erythropoietin2, VEGF3, HO14 and i-NOS5; all of which are involved in

promoting systemic O2 absorption and transportation6. However, these genes exert their effects

through distant effecter cells and tissues, such as EC and hematopoietic stem cells. Therefore,

they do little to restore the O2 homeostasis of tissue engineered constructs when cultured in

vitro. The reduction of ATP production rate causes the cytoplasm to accumulate ions that are

normally expelled via the cation pumping ATPase and eventually contributes to CM necrosis7.

Page 127: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

115

The most effective way to correct hypoxia is to increase the flux of O2 diffusion into the

engineered cardiac construct. This could be achieved by decreasing the average distance of CM

from culture medium and by increasing the O2 tension of culture medium. As covered in

previous chapters, numerous studies have attempted to vascularize tissue engineering constructs

with the view of reducing the distance of embedded cell from the culture medium. A recent

study published by the Sefton group indicates that the viability of embedded HepG2 cells can be

maintained in small diameter collagen modules, with a post-contraction diameter of 400 µm and

cell density of up to 8x107 cells/mL8. At the same time, HpeG2 cells embedded in large

diameter modules (700 µm, post-contraction) developed a necrotic core. In comparison, CM

embedded in similar size modules displayed core cell death at a cell density of 2x107 cells/mL,

which is likely due to their higher oxygen demand compared to HepG2 cells. Nevertheless,

these results suggest that the maximum density of viability cells supported by the modular

construct is inversely proportional to the diameter of the modules. For cardiac module, this may

represent a further reduction in module diameters, perhaps down to less than 100µm from the

current 200-300µm range, in order to maintain core viability at the current cell density.

Reduction of module diameter may be an adequate solution for improving O2 diffusion

in applications where the modules are meant to be used in their unassembled form, such as the

injection implant model. One of the potential issues when trying to create higher order structures

with smaller modules compared to larger ones is the increased number of intermodular junctions

per unit volume in the final assembled constructs. Assuming that these intermodular junctions

are mechanically weaker than the modules themselves, this would translate into an overall

decrease in mechanical strength of the assembled constructs. Indeed, during physical handling,

we found that the intermodular junctions in cardiac modular sheets tended to be the weaker

spots. Histological data suggested that cells in one module do not assemble into muscle bundles

with other cells in neighbouring modules. Therefore, increasing the number of intermodular

junctions using the current generation of cardiac modules will further decrease the mechanical

strength of the assembled constructs.

Other ways to increase O2 transfer into modules include tissue perfusion9 and increasing

O2 tension in the culture medium. Both methods operate by maintaining the concentration

gradient of oxygen across the module-medium interface. The amount of oxygen carried by

culture medium at saturation is governed by temperature and the partial pressure of O2 in the gas

phase above the liquid. In contrast, most of the oxygen carrying capacity of blood is handled by

Page 128: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

116

haemoglobin. Specifically, the total concentration of O2 from plasma and haemoglobin in

arterial blood can reach 8600 µM compared to 220 µM found in standard cell culture medium.

To compensate for the lack of haemoglobin, synthetic oxygen carriers have been formulated as

substitutes for haemoglobin in vitro10,11. Radisc et al. have found that the addition of

perfluorocarbon (PFC) emulsion in culture medium can increase the total amount of oxygen

delivery in a perfused cardiac bioreactor12. A similar strategy could be used to alleviate hypoxia

in the modular cardiac constructs.

5.2.2 Using autologous cell source

One of the crucial factors in successfully creating functional engineered tissue is to

select the correct cell source that complements the tissue engineering strategy. In this study, we

used neonatal rat CM as a model cell type to demonstrate the usefulness of the modular

construct. However, their allogeneic nature and scarcity in clinical settings severely limit their

potential as a viable treatment option for human patients. Recent advances in stems cell research

have gathered much attention as they hold the promise of generating the various cell types that

are necessary for cardiac tissue engineering with minimal immune barrier. For example,

autologous adult stromal stem cells can be readily harvested from patients’ own skin, fat, blood

or bone marrow and expanded ex vivo. Among these cell types, adipose stem cells (ASC) and

bone marrow derived mesenchymal stromal cells (bmMSC) are of particular interest. When

implanted into the myocardium, they have been shown to differentiate into myocytes and EC

respectively; two major cell types that are necessary to create cardiac tissue. In the context of

modular tissue engineering, ASC can be embedded into modules in their undifferentiated

progenitor state and allowed to mature in vitro into functional CM using biochemical factors,

including TGF-β113, VEGF14 and CM extracts15,16. Work by Tandon et al. demonstrated that

differentiated ASC respond to external electrical stimulation by upregulating connexin-43

expression as well as aligning and elongating perpendicular to the electric field17. Moreover,

ASC have been shown to differentiate into CM when injected directly into the myocardium18,19.

Together, these studies illustrate the potential of adult derived stem cell for generating

functional cardiac tissues.

Page 129: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

117

5.2.3 Characterization of host-module coupling

In order to maximize the functional benefits of modular cardiac tissue in the host’s

myocardium, the interface between host tissue and implanted modules must be electrically and

mechanically coupled with each other. Mechanical integration can be assessed by staining for

adherens junction components, including cardherins and desmosomes. Intermodular junction

must also be mechanically coupled to transmit force over the entire assembled construct. This

could be assessed by measuring the twitch force of the modular sheets in vitro. Ultimately, the

establishment of mechanical coupling allows the contractile forces generated by the implant to

be effectively transmitted to the myocardium.

The synchronous contraction of myocardium is achieved by the rapid and anisotropic

propagation of action potentials (AP) across the myocardium. This is mediated by gap junction

proteins known as connexins that are present on the surface of CM. Although the expressions of

connexins can be detected by immunohistological methods, only functional assays can confirm

that the connexins are organized into hemi-channels and that they are aligned with other hemi-

channels on neighbouring CM. The physical establishment of intercellular gap junction can be

detected using dye-transfer assay20. However, the diffusion of dye from one cell to another is

generally a much slower process compared to the propagation of AP. Therefore, the data

obtained will not carry sufficient spatial-temporal resolution to fully describe AP propagation on

the myocardium.

A more physiologically relevant assay to determine the functional status of gap junctions

is to optically or electrically measure the propagation velocity of AP across the modular sheet.

Changes in membrane potential during depolarization can be detected directly using ANEPPS

dyes, a family of fluorescence membrane dyes that varies their fluorescence output relative to

membrane potential21. Alternatively, CM depolarization can be detected indirectly by

monitoring intracellular calcium concentration using calcium sensitive fluorescent dyes. The

changes in fluorescence output in both systems can be mapped optically to indicate the

propagating front of AP across the modular sheet as a mean to assess the status of electrical

coupling between modules21,22. This method may also be applied to study electrical coupling

between the modular cardiac tissue and the host myocardium in explanted hearts. A similar

approach was used by Dumas et al., where the AP on a rabbit heart mounted in a Langendorff

perfusion apparatus was monitored using two-photon excitation of di-4-ANEPPS dye.

Page 130: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

118

Optical mapping of AP propagation across the cardiac modular sheets were attempted in

preliminary studies, but have achieved limited success. Upon further analysis, we have

identified several issues that may have hindered our ability to observe AP propagation across the

cardiac modular sheet. First and for most, the plane of AP propagation must be parallel to the

focus plane of the microscope in order to achieve accurate ratiometric fluorescence

measurements. This is mainly due to the drastic fall off of fluorescence signals in out-of-focus

planes. Although, this could be easily achieved in 2-D cultures, the curvatures of individual

module as well as the modular sheet have made it difficult to fit the entire depth of the modular

sheet within the focal plane. One way to correct this would be to mechanically fix the sheet on a

substrate, such as PDMS. Another solution would be to decrease the physical size of the

aperture of the lens in order to increase its depth of view, thereby increasing the depth of tissue

that would be in focus. However, in doing so, the number of photons reaching the camera would

be reduced and either the exposure times or camera sensitivity would have to be increased to

compensate for the decrease in signal. Of course, AP propagation could also be mapped using

an electrode array, whereby an electrical pulse is sent out to the test tissue by an input electrode.

The resulting changes in membrane potential are recorded using an array of recording electrodes

and the propagation velocity of AP is calculated based on the time delay and location of the

recorded signals. Since this modality relies on physical contact, it should solve some of the

issues being faced in optically mapping AP on modular cardiac tissues.

Results from these experiments could help elucidate the fate of modular cardiac

constructs in several ways. First, the electrophysiological properties of the implants can be

characterized. Studies have shown that engineered cardiac construct may be susceptible to

arrhythmia due to incompatible electrophysiological characteristics21,23. By mapping AP

propagation, any beneficial or adverse effects caused by the implant on the electrical conduction

in the host’s myocardium can be detected and studied. Overall, these data would provide

insights into the remodelling process that occurs during host-implant coupling.

Page 131: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

119

5.2.4 Assessing the functional benefits of modular cardiac tissue

The ultimate goal of engineered cardiac tissues is to replace and restore lost cardiac

function. Animal injury models like the ones described in this thesis serve as a useful platform

to assess the functional benefit of implanted tissue. It is necessary to quantify the functional

benefit of these implants in order to evaluate their efficacies and improve upon their designs.

From a clinical stand point, cardiac function refers primarily to the ability of the heart to pump a

sufficient volume of blood at the necessary pressure over each cycle. This can be translated to

measurable parameters, including end-systolic and end-diastolic ventricular pressures, ejection-

filling interval, stroke volume and ejection fraction. Many of these parameters can be

determined directly using a pressure-volume catheter inserted into the left ventricle24, or

measured using echocardiography as a non-invasive alternative.

Functional assays such as the one described above would be well suited for detecting the

restoration of function after implantation in an animal injury model, such as the peri-infarct

module injection model presented in this thesis. Moreover, control groups must be carefully

selected in order to obtain meaningful results. For example, in the module injection model, there

should be three control groups; one with CM-only modules, one with cell-free modules, and one

sham (saline injection). The differential changes in functional parameter between each group

would yield clues regarding the importance of each component in restoring cardiac functions.

These results would provide critical feedback for improving the therapeutic benefits of modular

cardiac tissues.

Page 132: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

120

Reference List

1. T. G. Smith, P. A. Robbins, and P. J. Ratcliffe, The human side of hypoxia-

inducible factor. Br. J. Haematol. 141, 325-334 (2008).

2. G. L. Semenza, M. K. Nejfelt, S. M. Chi, and S. E. Antonarakis, Hypoxia-

inducible nuclear factors bind to an enhancer element located 3' to the human

erythropoietin gene. Proc. Natl. Acad. Sci. U. S. A 88, 5680-5684 (1991).

3. J. A. Forsythe, B. H. Jiang, N. V. Iyer, F. Agani, S. W. Leung, R. D. Koos, and G.

L. Semenza, Activation of vascular endothelial growth factor gene transcription

by hypoxia-inducible factor 1. Mol. Cell Biol. 16, 4604-4613 (1996).

4. P. J. Lee, B. H. Jiang, B. Y. Chin, N. V. Iyer, J. Alam, G. L. Semenza, and A. M.

Choi, Hypoxia-inducible factor-1 mediates transcriptional activation of the heme

oxygenase-1 gene in response to hypoxia. J. Biol. Chem. 272, 5375-5381

(1997).

5. L. A. Palmer, G. L. Semenza, M. H. Stoler, and R. A. Johns, Hypoxia induces

type II NOS gene expression in pulmonary artery endothelial cells via HIF-1. Am.

J. Physiol 274, L212-L219 (1998).

6. G. L. Semenza, Hypoxia-inducible factor 1: master regulator of O2 homeostasis.

Curr. Opin. Genet. Dev. 8, 588-594 (1998).

7. M. Okuda, A multidisciplinary overview of cardiogenic shock. Shock 25, 557-570

(2006).

8. L. Corstorphine and M. V. Sefton, Effectiveness factor and diffusion limitations in

collagen gel modules containing HepG2 cells. J. Tissue Eng Regen. Med.

(2010).

9. M. A. Brown, R. K. Iyer, and M. Radisic, Pulsatile perfusion bioreactor for

cardiac tissue engineering. Biotechnol. Prog. 24, 907-920 (2008).

Page 133: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

121

10. F. T. Barbosa, M. J. Juca, A. A. Castro, J. L. Duarte, and L. T. Barbosa, Artificial

oxygen carriers as a possible alternative to red cells in clinical practice. Sao

Paulo Med. J. 127, 97-100 (2009).

11. T. Henkel-Honke and M. Oleck, Artificial oxygen carriers: a current review.

AANA. J. 75, 205-211 (2007).

12. M. Radisic, H. Park, F. Chen, J. E. Salazar-Lazzaro, Y. Wang, R. Dennis, R.

Langer, L. E. Freed, and G. Vunjak-Novakovic, Biomimetic approach to cardiac

tissue engineering: oxygen carriers and channeled scaffolds. Tissue Eng 12,

2077-2091 (2006).

13. S. J. Gwak, S. H. Bhang, H. S. Yang, S. S. Kim, D. H. Lee, S. H. Lee, and B. S.

Kim, In vitro cardiomyogenic differentiation of adipose-derived stromal cells

using transforming growth factor-beta1. Cell Biochem. Funct. 27, 148-154

(2009).

14. Y. H. Song, S. Gehmert, S. Sadat, K. Pinkernell, X. Bai, N. Matthias, and E. Alt,

VEGF is critical for spontaneous differentiation of stem cells into

cardiomyocytes. Biochem. Biophys. Res. Commun. 354, 999-1003 (2007).

15. K. G. Gaustad, A. C. Boquest, B. E. Anderson, A. M. Gerdes, and P. Collas,

Differentiation of human adipose tissue stem cells using extracts of rat

cardiomyocytes. Biochem. Biophys. Res. Commun. 314, 420-427 (2004).

16. M. Peran, J. A. Marchal, E. Lopez, M. Jimenez-Navarro, H. Boulaiz, F.

Rodriguez-Serrano, E. Carrillo, G. Sanchez-Espin, E. de Teresa, D. Tosh, and

A. Aranega, Human cardiac tissue induces transdifferentiation of adult stem cells

towards cardiomyocytes. Cytotherapy. 12, 332-337 (2010).

17. N. Tandon, B. Goh, A. Marsano, P. H. Chao, C. Montouri-Sorrentino, J. Gimble,

and G. Vunjak-Novakovic, Alignment and elongation of human adipose-derived

stem cells in response to direct-current electrical stimulation. Conf. Proc. IEEE

Eng Med. Biol. Soc. 2009, 6517-6521 (2009).

Page 134: Fabrication of a 3-dimensional cardiac tissue using a ... · Fabrication of a 3-dimensional cardiac tissue using a modular tissue engineering approach Brendan Martin Pue-Bun Leung

122

18. Y. S. Choi, K. Matsuda, G. J. Dusting, W. A. Morrison, and R. J. Dilley,

Engineering cardiac tissue in vivo from human adipose-derived stem cells.

Biomaterials 31, 2236-2242 (2010).

19. M. Jumabay, T. Matsumoto, S. Yokoyama, K. Kano, Y. Kusumi, T. Masuko, M.

Mitsumata, S. Saito, A. Hirayama, H. Mugishima, and N. Fukuda,

Dedifferentiated fat cells convert to cardiomyocyte phenotype and repair

infarcted cardiac tissue in rats. J. Mol. Cell Cardiol. 47, 565-575 (2009).

20. K. Unuma, K. Shintani-Ishida, K. Tsushima, T. Shimosawa, T. Ueyama, M.

Kuwahara, and K. Yoshida, Connexin-43 redistribution and gap junction

activation during forced restraint protects against sudden arrhythmic death in

rats. Circ. J. 74, 1087-1095 (2010).

21. Y. Itabashi, S. Miyoshi, S. Yuasa, J. Fujita, T. Shimizu, T. Okano, K. Fukuda,

and S. Ogawa, Analysis of the electrophysiological properties and arrhythmias in

directly contacted skeletal and cardiac muscle cell sheets. Cardiovasc. Res. 67,

561-570 (2005).

22. M. Radisic, V. G. Fast, O. F. Sharifov, R. K. Iyer, H. Park, and G. Vunjak-

Novakovic, Optical Mapping of Impulse Propagation in Engineered Cardiac

Tissue. Tissue Eng Part A (2008).

23. N. Bursac, Y. Loo, K. Leong, and L. Tung, Novel anisotropic engineered cardiac

tissues: studies of electrical propagation. Biochem. Biophys. Res. Commun.

361, 847-853 (2007).

24. P. Pacher, T. Nagayama, P. Mukhopadhyay, S. Batkai, and D. A. Kass,

Measurement of cardiac function using pressure-volume conductance catheter

technique in mice and rats. Nat. Protoc. 3, 1422-1434 (2008).