212
LOCAL, STRUCTURAL AND SYSTEMIC CUES FOR VASCULAR TISSUE ENGINEERING By VIDHYA RAMASWAMY A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2017

© 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

LOCAL, STRUCTURAL AND SYSTEMIC CUES FOR VASCULAR TISSUE ENGINEERING

By

VIDHYA RAMASWAMY

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2017

Page 2: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

© 2017 Vidhya Ramaswamy

Page 3: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

To every little girl with a dream of a career in science and engineering

Page 4: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

4

ACKNOWLEDGMENTS

சிறு துளி பெரு பெள்ளம் Little drops make the mighty ocean

This humble contribution of mine to the ocean of science would not have been

possible without, the immense support from friends, family, colleagues and esteemed

mentors. I would first like to acknowledge my advisor, Dr. Josephine Allen, for taking a

chance with me and giving me this opportunity and freedom to expand my skills and

research methodologies. Her patient guidance and faith in me, has been instrumental in

shaping my doctoral research experience. I would also like to thank my committee

members, Dr. Antonio Webb, Dr. Jennifer Andrew, Dr. Christopher Batich and Dr.

Chelsey Simmons for the valuable interactions and insightful questions, that have

provided me the framework for developing this dissertation. I am also greatly thankful

for the constant support and guidance I have received from each of my committee

members to shape my career. I would also like to specially acknowledge Dr. Nancy

Ruzycki, for her continual support and encouragement throughout my tenure in

graduate school. I have knocked on her door on countless occasions seeking help, and

I have always left her office with hope, enthusiasm and sweet treats. I would also like to

acknowledge my lab alumni and current members who have helped greatly and

contributed valuably to my research projects: Taylor Repetto, Elliott Dirr, Krista Dulany,

Allison Goins and Dr. Agata Zupanska.

Any achievement of mine is the result of the great amount of sacrifice, patience

and hard work of my parents, K. Ramaswamy and R. Vijayalakshmi and grandparents,

R. Krishnamoorthy and K. Pattammal. It is their constant unwavering belief in me and

limitless affection, that has fueled my career so far. I would also like to acknowledge

Page 5: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

5

with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great

source of inspiration, a dear friend, philosopher and a wise guide. Without his immense

patience, guidance and encouragement, my dream of a career in science would not

have been possible. A special thanks to my cousin, Sumathy who has been a big

source of inspiration for work-life balance.

Last but not the least, I want to thank all my wonderful friends: Praveen, Shalini,

Koushik, Pavithra, Neha, Aparna, Meena and Laura for being my personal

cheerleaders, and listening to my endless woes about graduate school and life. I am

permanently indebted to their belief in me, constant encouragement and positivity.

Page 6: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

6

TABLE OF CONTENTS Page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES .......................................................................................................... 10

LIST OF FIGURES ........................................................................................................ 11

LIST OF ABBREVIATIONS ........................................................................................... 14

ABSTRACT ................................................................................................................... 19

CHAPTER

1 INTRODUCTION .................................................................................................... 21

Specific Aim 1- Nanoscale Local Cues ................................................................... 23

Specific Aim 2- Mesoscale Structural Cues ............................................................ 23 Sub-aim 1 ......................................................................................................... 24 Sub-aim 2 ......................................................................................................... 24

Specific Aim 3- Macroscale Systemic Cues ............................................................ 24

2 BACKGROUND ...................................................................................................... 25

Cardiovascular Diseases and Tissue Engineering .................................................. 25 Nanoscale Local Cues ............................................................................................ 29

Mesoscale Structural Cues ..................................................................................... 35 Small Diameter Vascular Grafts ....................................................................... 38 Vascular Stents ................................................................................................ 42

Macroscale Systemic Cues ..................................................................................... 46

3 NANOSCALE LOCAL CUES- DEVELOPMENT OF A NOVEL VEGFR2 AGONIST APTAMER ASSEMBLY ......................................................................... 53

Introduction ............................................................................................................. 53 Specific Aim 1 ......................................................................................................... 56

Materials and Methods............................................................................................ 56 Aptamer Design ................................................................................................ 56 Aptamer-Endothelial Cell Binding ..................................................................... 58 VEGFR2 Phosphorylation ................................................................................ 60

Signal Pathway Activation ................................................................................ 61 Endothelial Nitric Oxide Synthase Expression .................................................. 61 Capillary Tube Formation (Angiogenesis) ........................................................ 62 Chorioallantoic Membrane (CAM) Assay.......................................................... 63

Results and Discussion........................................................................................... 64 Summary ................................................................................................................ 74

Page 7: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

7

4 MESOSCALE STRUCTURAL CUES- PART 1- DEVELOPMENT OF A BIOMIMETIC, BILAYERED VASCULAR SCAFFOLD FOR TISSUE ENGINEERING SMALL DIAMETER BLOOD VESSELS ........................................ 76

Introduction ............................................................................................................. 76 Specific Aim 2 ......................................................................................................... 77

Sub-aim 1 ......................................................................................................... 77 Sub-aim 2 ......................................................................................................... 77

Materials and Methods............................................................................................ 81

Materials ........................................................................................................... 81 Solid Lumen Fabrication ................................................................................... 82 Fabrication of Electrospun Medial Layer .......................................................... 82

Glutaraldehyde Crosslinking ............................................................................. 83 FTIR Characterization ...................................................................................... 84 SEM Characterization ...................................................................................... 84

Contact Angle Measurement ............................................................................ 84 Uniaxial Tensile Testing ................................................................................... 85

Biocompatibility Characterization ..................................................................... 85 Cell culture ................................................................................................. 85 Scaffold preparation for cell studies ........................................................... 85

Cellular attachment .................................................................................... 86 Lactate dehydrogenase cytotoxicity assay ................................................. 86

Cell proliferation assessment ..................................................................... 87 Optimizing Bilayer Fabrication with Electrospun POC-PAA.............................. 88 Fabrication of Bilayered Construct ................................................................... 88

Mechanical Properties ...................................................................................... 89 Compliance ................................................................................................ 89

Circumferential hoop stress ....................................................................... 89 Statistical Analysis ............................................................................................ 90

Results and Discussion........................................................................................... 90

5 MESOSCALE STRUCTURAL CUES- PART 2- HEMOCOMPATIBILITY EVALUATION OF A NOVEL MG-CA-SR ALLOY FOR VASCULAR STENT APPLICATIONS .................................................................................................... 106

Introduction ........................................................................................................... 106 Materials and Methods.......................................................................................... 110

Alloy Synthesis and Sample Preparation ....................................................... 110

Cytotoxicity Evaluation ................................................................................... 110 Hemolysis ....................................................................................................... 112 Evaluation of Platelet Adhesion and Activation .............................................. 113 Whole Blood Clotting Assessment ................................................................. 114 Release of Monocytic Inflammatory Cytokines ............................................... 114

Statistical Analysis .......................................................................................... 115 Results And Discussion ........................................................................................ 115 Summary .............................................................................................................. 125

Page 8: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

8

6 MACROSCALE SYSTEMIC CUES- PART 1- EFFECT OF SIMULATED MICROGRAVITY ON DIFFERENTIATION AND DOWNSTREAM FUNCTION OF PORCINE BLOOD DERIVED ENDOTHELIAL PROGENITOR CELLS .......... 128

Introduction ........................................................................................................... 128 Specific Aim 3 ....................................................................................................... 131 Methods and Materials.......................................................................................... 133

Isolation of Peripheral Blood Mononuclear Cells from Porcine Blood ............. 133 Rotating Wall Vessel Bioreactor System ........................................................ 134

Microgravity Culture of PBMNCs .................................................................... 134 Differentiation, Colony Formation and Immunocytochemistry ........................ 135 Endothelial-Like Cell Migration and Proliferation ............................................ 135

Nitric Oxide and Prostaglandin Secretion ....................................................... 136 Clotting Kinetics .............................................................................................. 137 Statistical Analysis .......................................................................................... 137

Results And Discussion ........................................................................................ 137

7 MACROSCALE SYSTEMIC CUES- PART 2- EFFECT OF SIMULATED MICROGRAVITY ON FUNCTION OF HUMAN BLOOD DERIVED ENDOTHELIAL CELLS......................................................................................... 147

Introduction ........................................................................................................... 147

Materials and Methods.......................................................................................... 149 Isolation of Peripheral Blood Mononuclear Cells from Human Blood ............. 149

Culture and Differentiation of Endothelial Progenitor Cells ............................. 150

Immunocytochemical Assessment of Differentiated Endothelial Cells ........... 150

Simulated Microgravity Culture of HE-Like Cells ............................................ 151 Cell Proliferation Assessment ......................................................................... 152 Gene Expression ............................................................................................ 152

Endothelial Cell Secretory Function ............................................................... 153 Plasma Clotting Kinetics ................................................................................. 153

Statistical Analysis .......................................................................................... 154 Results and Discussion......................................................................................... 154 Summary .............................................................................................................. 166

8 SUMMARY, CONCLUSIONS AND FUTURE WORK ........................................... 171

Summary of Results.............................................................................................. 171 Conclusions .......................................................................................................... 177 Future Work .......................................................................................................... 178

APPENDIX: PROTEIN CROSSLINKING USING CINNAMALDEHYDE VAPORS ...... 180

Electrospinning Gelatin Nanofibers ....................................................................... 181 Cinnamaldehyde Crosslinking and Preliminary Stabilization Assessment ............ 181 FTIR Characterization ........................................................................................... 182 SEM Characterization ........................................................................................... 183

Page 9: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

9

LIST OF REFERENCES ............................................................................................. 184

BIOGRAPHICAL SKETCH .......................................................................................... 212

Page 10: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

10

LIST OF TABLES

Table page 3-1 Aptamer sequences and configuration ............................................................... 57

4-1 Average fiber diameter of Collagen:POC:Elastin fibers with different cross-linking times. ....................................................................................................... 95

4-2 Contact angles at the water – biomaterial interface ............................................ 96

4-3 Uniaxial tensile test. Assessment of tensile properties of the glutaraldehyde vapor crosslinked electrospun mat ..................................................................... 97

4-4 Circumferential mechanical testing of bilayered scaffolds compared to native tissue and ePTFE.. ........................................................................................... 103

A-1 CA cross-linking conditions and resistance to degradation .............................. 181

Page 11: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

11

LIST OF FIGURES

Figure page 1-1 Multi-scale cues presented to cells ..................................................................... 24

2-1 Cardiovascular diseases and tissue engineering strategies. .............................. 27

2-2 Different types of cues that can direct stem cell behavior for tissue engineering. ........................................................................................................ 28

2-3 A representative schematic of a single iteration of the SELEX process. ............ 32

2-4 Mechanism of aptamer binding to complex substrate. ........................................ 33

2-5 Schematic representation of cellular response to microstructural and physical properties of scaffold matrices. ........................................................................... 36

2-6 A schematic representation of the structure of native artery.. ............................. 42

2-7 Some representative bioreactor systems for tissue engineering. ....................... 48

2-8 Path of a particle or cell aggregate in a RWV bioreactor illustrating the net forces acting on the particle in rotation. .............................................................. 50

3-1 Aptamer structure. .............................................................................................. 58

3-2 Schematic representation of the proposed method of action of the divalent aptamer assembly. Binding to target and activation via dimerization, membrane bound VEGFR2. ............................................................................... 65

3-3 Aptamer-cell binding interactions.. ...................................................................... 66

3-4 Aptamer Induce VEGFR2 activation and downstream molecular events ........... 68

3-5 Functional assessment of VEGFR2 aptamer binding. ........................................ 72

3-6 Chorio allantoic membrane assay. ..................................................................... 73

4-1 Schematic representation of the bilayered vascular graft. .................................. 81

4-2 Glutaraldehyde vapor crosslinking.. .................................................................... 83

4-3 Synthesis of poly diol citrates. ............................................................................ 91

4-4 SEM characterization as spun collagen:POC:elastin fibers. ............................... 93

4-5 SEM characterization of all crosslinking conditions using glutaraldehyde vapor.. ................................................................................................................ 94

Page 12: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

12

4-6 ATR-FTIR spectra of collagen: POC: elastin crosslinked fibers. ......................... 95

4-7 Contact angle measurements. ............................................................................ 96

4-8 SEM images of cell attachment. hASMCs seeded onto collagen: POC: elastin fibers. ...................................................................................................... 98

4-9 Relative cytotoxicity measurements of collagen: POC: elastin with hASMCs and POC lumen with HUVECs. .......................................................................... 99

4-10 Proliferation kinetics. ........................................................................................ 100

4-10 SEM characterization of POC; POC-PAA bilayered construct. ......................... 101

4-12 SEM characterization of POC; collagen: POC: Elastin bilayered construct ...... 102

5-1 Evaluation of cytotoxicity. ................................................................................. 116

5-2 Hemolysis assessment. .................................................................................... 118

5-3 Whole Blood Clotting assessment.. .................................................................. 120

5-4 Platelet adhesion and activation. ...................................................................... 122

5-5 Release of inflammatory cytokines/chemokines by THP-1 cells reported as absorbance at 450 nm. ..................................................................................... 124

6-1 RWV bioreactor set-up. .................................................................................... 132

6-2 Differentiation and characterization of PE-like cells. ......................................... 139

6-3 Proliferative and migratory functions of simulated microgravity pre-exposed PE-like cells. ..................................................................................................... 141

6-4 Representative results of quantification of release of soluble factors. .............. 143

6-5 Representative result showing quantification of blood clotting studies. ............ 145

7-1 Bioreactor set-up and operation. ...................................................................... 151

7-2 Isolation of PBMNCs and differentiation into ECs. ............................................ 156

7-3 Proliferation kinetics ......................................................................................... 158

7-4 Gene expression. ............................................................................................. 160

7-5 Secretory functions of cells.. ............................................................................. 161

7-6 Plasma clotting kinetics. ................................................................................... 164

Page 13: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

13

A-1 FTIR spectra of cross linked fibers ................................................................... 182

A-2 SEM characterization of CA crosslinking. ......................................................... 183

Page 14: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

14

LIST OF ABBREVIATIONS

µg Microgravity

ACD Acid Citrate Dextrose

AHA American Heart Association

ANOVA Analysis of Variance

Apt80mer 80 Mer VEGFR2 Binding Aptamer Sequence

AptDIVALENT Divalent VEGFR2 Binding Aptamer Assembly

AptM80mer Modified Base 80 Mer VEGFR2 Binding Aptamer Sequence

ASTM American Society for Testing and Materials

BME Basement Membrane Extract

BMP-2 Bone Morphogenic Protein

BSA Bovine Serum Albumin

CA Cinnamaldehyde

CaCl2 Calcium Chloride

CAM Chorio Allantoic Membrane

CD 309 Kinase Insert Domain Receptor Or VEGFR2

CD 31 Cluster of Differentiation 31

cDNA Complementary DNA

CPC Circulating Progenitor Cells

CSC Circulating Stem Cells

CVD Cardiovascular Diseases

DACRON Polyethylene Terephthalate

DMEM Dulbecco's Minimum Essential Medium

DNA Deoxyribonucleic Acid

Page 15: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

15

dsDNA Double Strand DNA

E Elastic Modulus

EBM-2 Endothelial Basal Medium-2

EC Endothelial Cells

ECM Extracellular Matrix

EGM-2 Endothelial Growth Medium-2

ELISA Enzyme Linked Immunosorbent Assay

eNOS Endothelial Nitric Oxide Synthase

EPC Endothelial Progenitor Cells

ePTFE Expanded Polytetrafluoroethylene

FBS Fetal Bovine Serum

FDA Food and Drug Administration

FITC Fluorescein Isothiocyanate

FTIR Fourier Transform Infrared Spectroscopy

GAPDH Glyceraldehyde 3-Phosphate Dehydrogenase

HARV High Aspect Rotating Vessel

hASMCs Human Aortic Smooth Muscle Cells

hEGF-2 Human Epidermal Growth Factor

HE-like cells Human Endothelial-Like Cells

hFGF Human Fibroblast Growth Factor

HRP Horseradish Peroxidase

HSP70 Heat Shock 70 Kda Protein

HSPA4 Gene Encoding Heat Shock 70 Kda Protein 4

HUVECs Human Umbilical Vein Endothelial Cells

IGF Insulin-Like Growth Factor

Page 16: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

16

IgG Immunoglobulin G

IL-1β Interleukin 1 Beta

IL-6 Interleukin 6

IL-8 Interleukin 8

ISO International Organization for Standardization

ISS International Space Station

LDH Lactate Dehydrogenase

LED Light Emitting Diode

MTT 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide

NASA National Aeronautics and Space Administration

NO Nitric Oxide

PAA Poly Acrylic Acid

PAEC Porcine Aortic Endothelial Cells

PBMNCs Peripheral Blood Mononuclear Cells

PBS Phosphate Buffered Saline

Pburst Burst Pressure

PCL Polycaprolactone

PCR Polymerase Chain Reaction

PDC Poly Diol Citrates

PDGF-BB Platelet Derived Growth Factor BB

PE Phycoerythrin

PEG Polyethylene Glycol

PE-Like cells Porcine Endothelial-Like Cells

PF 4 Platelet Factor 4

PGA Polyglycolic Acid

Page 17: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

17

PGE2 Prostaglandin E2

PLGA Poly (Lactic-Co-Glycolic Acid)

PLLA Poly (L-Lactic Acid)

POC Poly (1,8 Octane Diol-Co-Citrate)

PRP Platelet Rich Plasma

PU Polyurethane

PVDF Polyvinylidene Fluoride

qRT-PCR Quantitative Reverse Transcriptase - Polymerase Chain Reaction

r Radius of tubular scaffold

R3-IGF-1 Human Recombinant Insulin-Like Growth Factor

RBC Red Blood Cells

RNA Ribonucleic Acid

RWV Rotating Wall Vessel

S.S Stainless Steel

SELEX Systematic Evolution of Ligands by Exponential Enrichment

SEM Scanning Electron Microscopy

SMC Smooth Muscle Cells

STLV Slow Turning Lateral Vessel

t Thickness of tubular scaffold

TCP Tissue Culture Plastic

THP-1 Human Monocytic Cell Line

TNF-α Tumor Necrosis Factor Alpha

tPA Tissue Plasminogen Activator

VEGF Vascular Endothelial Growth Factor

VEGFR2 Vascular Endothelial Growth Factor Receptor-2

Page 18: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

18

vWF Von Willebrand Factor

ε Strain

σ Stress

Page 19: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

19

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

LOCAL, STRUCTURAL AND SYSTEMIC CUES FOR VASCULAR TISSUE

ENGINEERING

By

Vidhya Ramaswamy

August 2017

Chair: Josephine Allen Major: Materials Science and Engineering

The chronic scarcity of autologous donor organs to treat cardiovascular diseases

has led to increased efforts in alternate therapeutic strategies. One such approach is

the development of novel tissue engineering methods, to create functional replacements

for diseased or damaged parts of the vasculature. Living cells alter their behavior as a

direct response to intrinsic and extrinsic cues presented to them. The underlying

principle of tissue engineering is that the living cell system is the only true “tissue

engineer,” and the goal is to deliver appropriate cues to modulate the cellular response.

The goal of this dissertation is to study the underlying mechanism of cell-material

interactions at different scales to develop and deliver novel biomaterial based cues to

repair, preserve, and regenerate vascular tissue. Starting at the nanoscale, the first part

of this work involved the development of a novel divalent aptamer assembly that acts as

an agonist towards vascular endothelial growth factor receptor-2 in endothelial cells.

Besides activation, the assembly was also able to cause a positive downstream

angiogenic response, which proves that this is a promising strategy for vascular tissue

engineering. The second part of this work consisted of studying cues designed at the

meso scale including interaction of cells with surrounding scaffolds. This section

Page 20: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

20

covered two classes of cardiovascular biomaterials; small diameter vascular grafts and

vascular stents. The interaction of these biomaterials with in vitro models of the vascular

environment (blood and vascular cells) was studied to develop scaffold material based

strategies for tissue engineering. The third and final part of this dissertation was focused

on macroscale systemic cues, that provide a unique environment for modulating the

response of vascular cells. One such specialized environment is simulated microgravity,

which can be used to effectively study tissue engineering of 3D structures. The goal of

this work was to understand the behavior of circulating stem cells in this low shear

stress environment, to assess their therapeutic potential for vascular repair and

regeneration. Overall, this work is expected to contribute towards widening the

knowledge base in the study of cell-material interactions and open avenues for research

in this field.

Page 21: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

21

CHAPTER 1 INTRODUCTION

Despite remarkable advances in research in the management and prevention of

complications arising from cardiovascular disease (CVD), it still remains the leading

cause of death worldwide1–3. In the United States alone, nearly $300 billion is spent on

healthcare costs annually towards managing CVD4. In most cases, these diseases

cause irreversible organ damage where the body is not able to regenerate or repair the

damage by itself. These complications have created a crucial need for novel methods to

stimulate repair and regeneration of the cardiovascular system. Traditional treatments

for CVD include medications and surgical interventions. However, most surgical options

involve circumventing the diseased or damaged vascular tissue, (bypass stents or

grafts), or completely replacing them (transplants). But the chronic shortage of available

donors, has steered the treatment of these conditions towards developing alternative

therapies such as tissue engineering, which can significantly expand patient care

options. The goal of such strategies is to efficiently provide means for repair,

preservation and regeneration of vasculature to establish normal functioning of the

cardiovascular system4,5 .This approach will also greatly diminish the need for scarce

donor organs, thereby improving clinical point of care for patients affected by these

diseases. This need is further fueled by aging population and increased expectations for

a better quality of life. Besides their intended use, the new engineered tissues also find

application in disease studies, drug testing and in vitro development organotypic model

systems6,7.

Vascular tissue engineering is aimed at applying principles of engineering and

life sciences towards the development of novel strategies to restore, maintain and

Page 22: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

22

enhance cardiovascular tissue function6,8,9. Living cells react and alter their behavior as

a direct response to intrinsic and extrinsic cues presented to them. This behavior makes

it possible to design vehicles to deliver these cues to cell systems10. The use of

biomaterials as potential tools to achieve these end goals has been gaining popularity

recently. This is mainly due to the flexibility to alter physical, chemical and structural

properties of biomaterials to invoke a specific cellular response11. These cues can be a

variety of different factors such as pH, temperature, pressure, nutrient supply and waste

removal, mechanical, electrical and magnetic stimuli, etc. These in turn can cause the

modulation of important cellular functions such as cell growth, migration, differentiation,

survival or even apoptosis. These cues can also be applied across various length

scales varying from subcellular molecular cues to systemic macroscale cues12–14.

Biomaterials engineering exploits the structure-property relationship of

biomaterials to present these specific cues that can modulate a favorable response from

cell systems. This field makes use of a large and extensive toolkit comprising of material

parameters and properties available to modify or control an exact cellular function15,16.

Extensive research over the past few decades has shown that while designing

strategies to create living replacement for existing vascular tissue, the complexity of the

organ system itself must be taken into consideration. The underlying principle is that the

living cell system is the only true “tissue engineer,” and the goal is to deliver appropriate

stimuli, to the cellular microenvironment on multiple hierarchical levels to effectively

recreate tissue formation17,18.

Tissue formation in the body is a direct result of highly synergistic and

coordinated sequences of cell renewal, growth, differentiation, and assembly, within a

Page 23: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

23

dynamic environment characterized by spatial and temporal gradients of multiple

factors6,8,17. Therefore, to replicate and stimulate these functions of cells, it is imperative

to study and understand the biological stimuli at multiple levels to design and deliver the

appropriate cues. Thus, there is a need for a multifaceted approach spread across

different scales while studying strategies to enhance tissue engineering. The goal of this

dissertation is to study the underlying mechanism of cell-material interactions at

different scales (nano, meso and macroscales), to develop novel strategies to repair,

preserve and regenerate vascular tissue. A thorough and comprehensive understanding

of the complex and vast mechanisms of complete tissue regeneration is challenging and

is out of the scope of this work. However, this study provides essential elements of

understanding, of distinct systems towards furthering the knowledge in the field of

vascular tissue engineering. The dissertation has been divided into three parts with

each part discussing studies dealing with a specific length scale as shown in Figure 1-1

below. To probe cells at each of the length scales of interest, I have established the

following three specific aims pertaining to each specific scale of interest.

Specific Aim 1- Nanoscale Local Cues

Design and develop of a novel divalent DNA oligonucleotide sequence (aptamer),

that when assembled into a unique divalent assembly acts as a receptor agonist towards

VEGFR2.

Specific Aim 2- Mesoscale Structural Cues

Understand and develop biomaterial based strategies towards vascular tissue

engineering and regeneration by studying the interactions of biomaterials with in vitro

vascular microenvironments.

Page 24: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

24

Sub-aim 1

Develop a novel biomimetic, bilayered, biomaterial construct and evaluate its

applicability as a small diameter vascular graft.

Sub-aim 2

Evaluate the biocompatibility and hemocompatibility of an Mg-Ca-Sr alloy to

assess it suitability for use in designing vascular stents.

Specific Aim 3- Macroscale Systemic Cues

Study and understand the effect of simulated microgravity on the function of

vascular stem cells to assess their potential to be used as a therapeutic strategy for

vascular repair and regeneration.

Figure 1-1. Multi-scale cues presented to cells

Page 25: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

25

CHAPTER 2 BACKGROUND

Cardiovascular Diseases and Tissue Engineering

In the statistical update summary released by the American Heart Association

(AHA), the overall rate of death attributed to Cardiovascular diseases (CVD) alone was

reported to be 235.5 per 100,000 in 20101,19. This data revealed that more than 2150

Americans die of CVD every day with an average of 1 death every 40 seconds. The

total cost of medical procedures performed in the treatment of CVD, amounts to over

300 billion dollars each year3,19,20. The treatment costs of CVD alone were more than

any other disease group. With these numbers growing every year, treatment strategies

to manage and alleviate the complications arising from CVD has become a national

concern.

Cardiovascular diseases can manifest in different ways in the body, often

resulting in death. Some of the conditions that can be classified under the broad

umbrella of cardiovascular diseases include rheumatic heart disease, congenital heart

disease, coronary heart disease, cardiomyopathy, deep vein thrombosis, inflammatory

heart disease and other disorders19. Although, significant efforts are taken towards

preventing the occurrence of these conditions by closely monitoring the underlying

causative factors, and providing additional care for patients in the high-risk category,

there has not been a significant reduction in mortality due to CVD. Therefore, a great

portion of healthcare costs are being invested in CVD treatment every year.

Current treatment strategies include, pharmaceutical drugs and novel medical

devices which have significantly improved the quality of life for patients with CVD. But

these have failed to help patients with end-stage CVD. Moreover, in some cases

Page 26: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

26

despite drug therapy the condition eventually becomes refractory to therapy and

disease progression is inevitable4,21. The golden standard for treatment of CVD has

been complete organ replacement or partial tissue transplantation. But this approach is

very much limited by the scarcity of suitable autologous tissue that is healthy and

available for transplantation. With the exponential increase in the need for donors which

is not matched by the availability of donors, more recent approaches have been focused

on developing tissue engineering based strategies to replace diseased and damaged

part of cardiovascular tissue4,6.

Tissue engineering is a broad field of science that combines the principles of

engineering, material science, and biology into development of functional tissue

substitutes that can repair, maintain, or regenerate tissue function in damaged

segments of the body6. Within the field of cardiovascular tissue engineering, a great

amount of progress has been made. Tissue engineered replacements for various

constituents of the cardiovascular system, including blood vessels, heart valves, and

cardiac muscle have been developed over the years4,8,22,23. The extent of progress

made in this field of research has created a promising environment for widespread

application of tissue-engineered therapy for CVD.

Many noteworthy studies have reported new and improved techniques including

endothelial cell seeding of vascular grafts, tissue-engineered vascular conduits,

generation of heart valve leaflets, cardiomyopathy, genetic manipulation, and in vitro

conditions for optimizing tissue-engineered cardiovascular constructs11,22–25. Many of

these cross-over concepts have been simultaneously applied to treat multiple disorders

of the heart. The figure below (Figure 2-1) summarizes some of the recent tissue

Page 27: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

27

engineering based therapy developed for various complications due to CVD. From the

tissue engineering point of view, the schematic below also highlights the importance of

using biomaterial scaffolds as delivery vehicles for appropriate vascular cells on the site

of injury, to enable regeneration. The figure also shows the need for diverse scaffold

designs as well cell types, used for different tissues in the cardiovascular system.

Figure 2-1. Cardiovascular diseases and tissue engineering strategies.Image reference :http://www.sciencecodex.com/files/different%20ways%20in%20which%20tissue%20engineers%20believe%20that%20heart%20muscle.jpg

The underlying concept behind tissue engineering therapy is that, the living cell

system is the ‘true tissue engineer’. Any tissue engineering strategy is focused on

exploiting the resilient ability of this system to recuperate and regenerate damaged

tissue by modulating its response when provided with the appropriate stimuli6,17,26.

Page 28: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

28

Living cells are highly sensitive to extrinsic cues that can control their functions.

Their complex sensory mechanisms can detect and respond to a broad variety of

signals that can be transduced into useful cell functions10,13,17,18. These cues can range

from environmental changes such as change in osmolarity, pH, temperature, etc., to

subcellular molecules such as growth factors, cytokines, etc. These in turn play a very

important role in regulating important cell functions, such as cell migration,

differentiation, proliferation and even apoptosis. One such example is represented in

Figure 2-2 below, showing a schematic summarizing different types of cues that can be

applied to a stem cell environment to modulate a favorable response for tissue

engineering.

Figure 2-2. Different types of cues that can direct stem cell behavior for tissue engineering. Image reference: Mashayekhan S, Hajiabbas M, Fallah A. Stem Cells in Tissue Engineering. In: Pluripotent Stem Cells. Vol InTech; 2013. doi:10.5772/54371.

Page 29: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

29

Another important point to note is that, these extrinsic cues applied to cell

systems can either be employed separately, or in correlation with each other. These

cues can often work synergistically and generate a complex response otherwise

unachievable by modulating a single factor. This concept is represented by the

interconnected lines in the above schematic as well.

This sensitivity of living cell systems has been exploited in directing cell function

for tissue engineering and regeneration for many years now. Biomaterials engineers are

focused on designing appropriate cues to control cell functions for tissue engineering. In

the process of designing cues, the science behind the interaction of cell systems with

the external micro/macro environment has also been extensively studied and

reported10,26,27. Many significant contributions to this field have been made in the recent

past that have revolutionized biomaterial design in tissue engineering. This chapter will

briefly cover the major trends in technological advancement in this field. For better

organization and understanding, the following text has been divided into 3 subsections,

including discussion about specific cues at 3 different scales (nano, meso and macro).

The rationale behind this organization is to correlate this brief review with the theme of

this dissertation as described in the Chapter 1.

Nanoscale Local Cues

From the birth of nanotechnology in 1959, through Richard Feynman’s seminal

lecture, this field has metamorphosed into an all-encompassing technology applied

across the scientific realm28. The ability to precisely tailor bulk properties of materials by

hierarchical design of nanomaterial constituents has caused increased interest in this

field. In addition to design of materials, the vested interest in nanotechnology has also

caused studies into understanding phenomenon happening at the nanoscale.

Page 30: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

30

Application of nanotechnology to tissue engineering and regeneration, stems

from the hierarchical nanostructure of cells and tissues and the extracellular

environment29,30. Initial attempts at tissue engineering failed to consider the dynamic

microenvironment of the cells and its effect on altering cell behavior. As interest grew in

this field, emphasis was placed on mimicking the extracellular environment in healthy

tissues to regenerate and repair damaged tissues10,31. Instead of simply delivering cells

in the diseased area, this approach was combined with stimuli that help engineer the

cells to repair and regenerate the tissue. These cues vary both in scale and

spatiotemporal functionality13.

To truly mimic the extracellular environment, it is essential to start from the nano

or subcellular scale. Many studies over the years have revealed the potential

importance of cues at the nanoscale to affect important functions of cells such as

signaling, migration, adhesion, proliferation, and differentiation18,30. Various strategies to

deliver nanoscale cues to cells have since been investigated. Some current strategies

include but are not limited to, nanopatterned biomaterial scaffolds, nanoparticles,

nanodevices, proteins, growth factors, chemokines and small molecules18,30,32,33.

In particular the delivery of biological and synthetic signaling molecules such as

low-molecular-weight drugs, proteins and oligonucleotides that can stimulate cell

migration, growth and differentiation for tissue engineering has been successfully tried

and tested in in vitro settings34–36. The design strategies of signaling molecules has

been based on the understanding of natural tissue morphogenic mechanisms which is a

coordinated action of multiple signaling molecules. Broadly, two categories of such

molecules have been studied in relation to tissue engineering therapy; 1) soluble

Page 31: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

31

signaling molecules such as growth factors and other regulatory proteins that are

secreted within the cell system’ and 2) Insoluble signaling factors derived from the ECM

environment. Of these, the delivery of many growth factors such as vascular endothelial

growth factor (VEGF) which regulates and controls primarily all vascular repair and

angiogenic mechanisms, platelet derived growth factor (PDGF-ββ) which promotes

would healing in most tissues, bone morphogenic protein-2 (BMP-2) which has known

to improve bone regeneration mechanisms have been studied extensively34,37,38.

Although many of these growth factors have been used in clinical trials for tissue

regeneration therapy, only PDGF- ββ, BMP-2 and BMP-7 have shown promise37,39–42.

These factors however are not cost effective and often have safety limitations. For

example, despite successful preclinical and phase I studies, clinical trials of VEGF

therapy have not been very encouraging. Particularly the delivery of VEGF at safe

dosages to target tissues to induce tissue repair has been a major challenge37,43,44.

Other engineering challenges for delivering growth factors include the need to control

the microenvironmental tissue distribution, which is inherently difficult with gene delivery

techniques. Although delivery of recombinant proteins could overcome the distribution

issue, they have too short half-lives in vivo. Additionally, growth factors also cause non-

specific activation events and invoke immunogenic responses which are not desirable45.

Therefore, current work in this field has been focused on alternate synthetic molecules

and engineered signaling factors that can specifically target cellular domains and

stimulate tissue regeneration. A promising approach to this challenge is the delivery of

synthetic peptides and engineered RNA and DNA sequences, such as aptamers, to

Page 32: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

32

target and activate specific targets within the cells35,43,46. Some preliminary studies have

shown encouraging results via the use of aptamers towards this goal47,48.

Nucleic acid aptamers are short single-stranded oligonucleotides which bind to a

pre-determined target with very high specificity and binding affinity. Aptamers were first

discovered nearly 3 decades ago, in 1990, when several groups are attributed to have

isolated RNA aptamers. In the pivotal work by Tuerk and Gold where two high affinity

RNA ligands were enriched from a randomized library49. The iterative selection method

used for enriching specific ligands was named as SELEX, short for systematic evolution

of ligands by exponential enrichment. In a separate work by Ellington and Szostak, the

term aptamer was coined to refer to high affinity molecules35. A representative

schematic explaining the SELEX process is shown below in Figure 2-3. The figure

shows a single iteration process in SELEX where high affinity aptamers are selected

through a sequentially purifying candidate sequences by binding to a target molecule.

Figure 2-3. A representative schematic of a single iteration of the SELEX process.

Page 33: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

33

The group was able to identify and isolate RNA aptamers that could specifically

bind to organic dyes. Ever since their initial discovery and isolation, thousands of

aptamers have been isolated for a variety of targets including but not limited to peptides,

proteins, viruses, bacteria, receptors, organic molecules, metallic ions and even whole

cells47,48,50–53.

Aptamers have a unique advantage when compared to other oligonucleotides

such as siRNAs in therapeutics. While siRNAs can only be designed to intracellular

targets, aptamers are more flexible and can be developed to target cell-surface, intra

and extracellular substrates54,55. The therapeutic ability of aptamers have been largely

based on their ability to form complex 3D secondary structures such as hairpins, loops,

quadruplexes etc. that enables them to dock on and bind to complex domains on

substrates51. The mechanism of specific binding of aptamers to substrates on cells is

represented in the schematic below.

Figure 2-4. Mechanism of aptamer binding to complex substrate.

This characteristic of aptamers has been largely used to inhibit protein-protein

interactions such as receptor-ligand interactions thereby acting as antagonists of

receptor function. Although the binding affinity and specificity of aptamers make them

ideal therapeutic agents, in vivo translation and delivery of aptamers has proven to be a

Page 34: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

34

challenge. The clinical studies for the use of aptamers have been largely limited by their

susceptibility to nuclease-mediated degradation in the presence of serum and renal

filtration. This reduces their circulation time and affects their efficacy in the body.

However, more recent studies showed that this challenge can be overcome by

chemically modifying the nucleotide sugars or internucleotide phosphodiester linkages

55. Further, these modified aptamers have also been screened using the SELEX

process. Another promising approach has been the capping of the oligonucleotide

termini, thereby increasing its stability in serum. With these important advancements,

many aptamer ligands have been identified for variety of targets such as Angiopoietin1,

Angiopoietin 2, VEGF, Nucleolin, Thrombin, PDGF etc47,53. Of which many aptamers

such as Pegaptanib (Targeting VEGF), AS1411 (targeting nucleolin in cancer cells),

NU172 (targeting thrombin) etc. have been approved for use clinically as well 48,56–60.

With increased advancement in the therapeutic applications of aptamers which

exploit their antagonistic ability, their potential application in tissue engineering and

regeneration has also been investigated. Aptamers are being increasingly used to

capture cells and proteins on biomaterial scaffolds to promote cell infiltration and

endothelization of scaffolds. Additionally, a new and exciting field of research with

aptamers has been their use to modulate cell function for tissue engineering

applications. Remarkable work by Dollins et al.61, reported the synthesis of an OX-40

specific aptamer assembly that acted as a receptor agonist. Since then there has been

increased interest in developing aptamer based agonists. A recent report has also

identified an aptamer that can cause activation of insulin receptors62. This work

describes the design and development of a divalent aptamer assembly for controlled

Page 35: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

35

activation of VEGFR2 receptor in endothelial cells in Chapter 3. This study opens

avenues to develop aptamer based strategies to control and modulate cellular functions

for tissue engineering applications. Due to the flexibility of designing and developing

aptamer substrates and the wide variety of target molecules that they can be designed

for, a potential direction for future work in this field, is developing supra-molecular

multimer assembly that can deliver specific subcellular cues to cells to program their

function to repair and regenerate tissue in vivo60.

Mesoscale Structural Cues

The previous section discussed the importance of designing stimuli at the

nanoscale. Especially the use of small molecule triggers to control and modulate cellular

function. Most tissue systems are known to be anchorage dependent which means that

they require a substrate to attach to and grow. In healthy tissues this support is provided

by the extra cellular matrix that surrounds the cellular environment.

As discussed earlier, the extra cellular environment is a complex hierarchical

system constituting of multiple levels of functional components. This complex structure

makes it essential to study the cues presented by each hierarchical level to mimic the

existing extracellular environment63. Moving up on the scale of hierarchy, this section

describes strategies to modulate cellular response by designing appropriate cues on the

mesoscale. The mesoscale is defined as being inclusive of constituents ranging

between 10 to 100 microns in size14. This scale includes the interactions between

biomaterial substrates and cells. These interactions can be divided broadly into two

categories, (a) cells that are in direct contact with biomaterials, and (b) cells that are

indirectly exposed to an environment which consists of biomaterials.

Page 36: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

36

Important cellular responses and characteristics including alteration of cell shape,

alignment, differentiation, growth and tissue formation have been known to be

modulated by architectural cues that are presented to the cell systems by biomaterials

that are in contact with them14,30,33. This concept has inspired the design of biomaterials

for tissue engineering. For example mesoscale structural parameters such as fibers,

grooves, patterns, roughness as well as physical cues such as stiffness, surface

chemistry, elasticity and micro hardness of substrates have known to play an important

role in cellular morphology, alignment, growth, migration and differentiation14,26. This is

depicted in the schematic shown in Figure 2-5 below. A typical example of this

phenomenon is the behavior of stem cells in substrates of varying stiffness. In the

seminal work by Adam Engler in 200664, the dependence of matrix stiffness in directing

stem cell lineage was reported. Similarly many studies since then have shown the effect

of substrate mechanical and topographic properties on stem cell fate65.

Figure 2-5. Schematic representation of cellular response to microstructural and physical properties of scaffold matrices. Image reference: Kollmannsberger P, Bidan CM, Dunlop JWC, et al. The physics of tissue patterning and extracellular matrix organisation: how cells join forces. Soft Matter. 2011;7(20):9549.

Page 37: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

37

In the vascular world, biomaterial substitutes have been used to engineer a

variety of scaffolds and devices that are used to treat many vascular diseases as shown

in Figure 2-1. Early vascular biomaterial substrates and devices were devised as a

means of support to damaged or diseased vasculature and to house transplanted

cells/tissues to repair and regenerate the area. However, over the years, the inherent

functionality and cues provided by these scaffolds have also gained importance.

Biomaterials used for vascular tissue engineering can broadly be classified into two

categories; synthetic and natural materials66,67. Synthetic materials used in vascular

tissue engineering for cardiovascular system primarily consists of polymers and metals

or sometimes even a composite of both. Natural materials are mostly derived from plant

or animal tissues. They are inherently the preferred choice for biomaterial implants due

to their excellent biological functionality that makes them acceptable for tissue

engineering applications. Natural materials have the advantage of possessing excellent

biocompatibility, being non-immunogenic and eliciting minimal inflammatory response

from the body. However, they do not match up to the expected levels of mechanical

robustness for a cardiovascular implant in most cases. Additionally they are also

expensive and difficult to process68,69. Therefore, synthetic materials have been

researched as an alternative. One of the major advantages of synthetic materials is the

ability to tailor their physical, chemical and topographical properties. They are stronger

and more mechanically robust in comparison to natural biomaterials. They also are

cheaper to obtain and possess more flexibility of processing. The application of

synthetic materials is however greatly limited by concerns of long term cytotoxicity and

inflammatory response by the body66,67,70,71.

Page 38: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

38

The definition of biocompatibility of biomaterials is dynamic and has been

changing with advancement in research in this field. However, the general requirements

of a cardiovascular biomaterial include, mechanical compatibility, low cytotoxicity,

noncalicification, compatibility with blood and blood components, minimal inflammatory

or immunogenic response and in most cases capability to support vascular cell

systems66,67. Traditionally, preferred biomaterials used for medical applications have

been inert systems that do not cause adverse reactions in the body. However more

recent research has been focused on developing functional bioactive scaffolds that can

deliver cues to cell systems that come in contact with them. The idea behind this

approach is to design and tailor the chemical, physical or topographic features of

biomaterial substrates to effectively influence the interaction at the cell-biomaterial

interface.

Cardiovascular biomaterials in general encompasses a wide variety of material

applications ranging from vascular grafts to hemodialysis tubes. These biomaterials can

be designed to alleviate complications arising due to different cardiovascular diseases,

such as atherosclerosis, myocardial infarction, peripheral artery disease etc. This

section will briefly discuss two classes of cardiovascular biomaterial applications,

namely, small diameter vascular grafts and vascular stents, respectively. This section

will also briefly summarize the interaction of these biomaterials with in vitro models of

the vascular environment to understand and develop strategies to repair damaged

vasculature and enable regeneration.

Small Diameter Vascular Grafts

Owing to the clinical difficulty in obtaining autograft blood vessels to replace

diseased vasculature, alternative strategies are much sought after. Early studies dating

Page 39: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

39

back to 1980’s reported the development of completely synthetic grafts25,72,73. These

studies led to the development two important candidate materials for vascular grafts;

polyethylene terephthalate (Dacron®; INVISTA Technologies S.à.r.l., Saint Gallen,

Switzerland) and expanded polytetrafluoroethylene (Teflon®; E.I. du Pont de Nemours

and Company, Wilmington, DE, USA). Both these materials have been used clinically,

with good success rates as replacement materials for large diameter blood vessels73,74.

However, they failed to perform adequately in small-diameter vessels with disappointing

results from clinical attempts. Many synthetic grafts since then have been developed but

they have proven to be largely unsuccessful resulting in low patency and clinical

complications such as neointimal hyperplasia and increased thrombosis71,75–77. As an

improvement on the previous materials, polyurethanes were then identified as potential

candidates for the vascular grafts. Polyurethanes exhibit good mechanical compliance

thromboresistive characteristics and support the growth of living tissue. However their

use was limited by their poor biostability71.

After the initial setbacks in the use of purely synthetic substrates, emerging

needs steered the research in this field from ’replacement’ to ‘regeneration’ of the

functional tissue. To this end, a biomimetic approach was employed while designing

scaffolds for regeneration. More and more natural materials were included in the

scaffold design. In an important work by Weinberg and Bell in 1986, collagen tubular

scaffolds seeded with cells was studied and this study became one of the first reports to

use protein based scaffolds78. Even though since then many collagen based scaffolds

have been fabricated and also have been utilized in animal studies, they lacked

mechanical strength and required the use of a synthetic support layer. Many other

Page 40: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

40

natural materials such as fibrin, silk, gelatin and elastin have been used to create

vascular grafts but all of them have had limited success due to mechanical mismatch

with native tissue68,79. Therefore, more recently there has been interest in developing

composite materials which are mechanically compatible but also possess the biological

functionality of natural materials. Natural ECM proteins such as collagen and elastin

have been blended with synthetic polymeric systems such as poly(l-lactide) (PLLA),

polycaprolactone (PCL), Poly(l-lactide-co-glycolide) (PLGA) and polyurethanes (PU).

The first part of the work discussed in Chapter 4 of this dissertation describes the

development of one such protein-polymer hybrid scaffold for small diameter vascular

grafts. This study investigates the use of a combination of a biodegradable, elastomeric

polymer; poly (1,8 octanediol-co-citrate) (POC) with natural extracellular matrix proteins;

collagen and elastin. POC belongs to a family of elastomers composed of citric acid and

an aliphatic diol. POC has been identified as a hydrophilic and biodegradable polymeric

constituent that can substitute for elastin in synthetic fibres80–83. It is also anti-

thrombogenic and supports differentiated endothelial progenitor cell adhesion and

growth84,85.

Even though the base material of the scaffold largely determines its physical,

chemical characteristics and its mechanical properties, the processing of scaffold plays

a major role in determining its functional ability. The scaffold must be fabricated in the

correct shape and size matching the target tissue, and be sufficiently strong to

withstand repetitive mechanical loading. The scaffold must also promote appropriate

vascular cell phenotype and present a large surface area to volume ratio to enhance

cell attachment and growth86. Further the scaffold design should include porous areas to

Page 41: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

41

improve cell migration, signaling and 3D vasculogenesis87.This porous network is

important for nutrient delivery, waste removal, and migration of cells and signaling

molecules33,66,88,89.

A biomimetically inspired scaffold design attempts to best represent the

architecture of the native ECM. Native artery as shown in Figure 2-6 consists of a solid

inner layer known as the lumen and a fibrous exterior layer also known as the medial

layer. The inner luminal layer is composed primarily of a monolayer of endothelial cells

that are in direct contact with circulating blood. The medial layer, consists of multiple

layer of smooth muscle cells. This layer is also rich in collagen and elastin among other

ECM proteins and provides a great amount of mechanical support to the vessel90–92.

This organizational structure is essential to enable vascularization and enhanced

reintegration into existing healthy vasculature. Therefore, some studies in the past

decade have adopted this concept and designed vascular scaffolds consisting of

multiple layers93–95. As mentioned above the scaffold design has also been made to

include porous layers to account for the 3D integration of cells and vascular growth.

Many techniques such as use of porogens, 3D printing, electrospinning, solvent casting

and phase separation has been employed to include porosity in scaffolds86,96,97. Of

these techniques, electrospinning offers robust material selection, low cost, simplicity,

high surface-to-volume ratios, and controlled porosity for vascular scaffold fabrication.

With these advantages, electrospinning is a proven framework allowing for the improved

endothelialization of vascular grafts. In this context, Chapter 3 describes the design and

development of a bilayered vascular graft that consists of a solid lumen layer and a

fibrous medial scaffold layer to mimic the native architecture of the blood vessel.

Page 42: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

42

Figure 2-6. A schematic representation of the structure of native artery. Image reference: Blausen.com staff (2014). "Medical gallery of Blausen Medical 2014". WikiJournal of Medicine 1 (2). DOI:10.15347/wjm/2014.010. ISSN 2002-4436.

Future work in this field is focused on identifying and utilizing an appropriate cell

source to promote the reendothelization of vascular grafts. Therefore, the next

generation grafts are being designed to recruit stem cells from circulating blood to mimic

natural blood vessel regeneration in the body. Additionally, further topographical

modulations, and functionalization of scaffold surface with bioactive molecules are being

investigated to improve graft functionality46,98–100.

Vascular Stents

Metals dominate the field of cardiovascular biomaterials next to if not equal to

polymeric materials67. The use of metals for biomedical application dates to as early as

1985. Majority of the applications of metals in medicine was in the form of heart valves,

Page 43: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

43

vascular stents and stent-graft combinations. The excellent mechanical properties and

workability of metals have furthered the application of metals for high stress applications

within the body101. However, some of the commonly used bio-metals possess limited

biofunctionalities such as blood compatibility, bone conductivity and bioactivity. To be a

candidate material for cardiovascular applications, a material must exhibit compatibility

with the cellular environment including blood and its components. Failure of which,

leads to complications such as thrombosis and inflammation which not only prevents

incorporation of the implant into the existing vasculature but also causes significant

damage to it.

Balloon-expandable stents were first used in 1985 for coronary arteries to treat

complications arising from coronary artery diseases24,67,102. Many metals have been

used to fabricate vascular stents since then. Some of the earlier stent design utilized

inert materials such as stainless steel. Further modifications were made to the

composition of the stainless steel such as addition of Cr and Ni to improve the strength

of the alloy. It was then furthered altered by reducing the percentage of carbon and

include molybdenum to improve corrosion resistance101,103. This composition was

designated as 316 L which is still currently the golden standard for stents and several

316 L SS have also been FDA approved for stent applications. Several other metals

such cobalt, tantalum, molybdenum, titanium and their alloys were also used to develop

metallic stents. Stents made of Ti and its alloys were particularly preferred for these

applications due to their light weight, high strength and excellent corrosion resistance.

Even though these bare metal stents performed well in terms of mechanical behavior,

they had serious setbacks such as increased thrombotic or inflammatory response.

Page 44: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

44

More importantly, due the permanent nature of these implants, they also caused

damage to vessel geometry and caused negative remodeling101,102,104. Therefore,

another important development in this field was the noteworthy discovery of degradable

stents that gradually dissolve away and are replaced in entirety by the native tissue.

These stents are designed to assist in arterial remodeling, a process which has been

showed to take 6-12 months105–108. Therefore, the degradation rate of materials chosen

for this application was designed to match this rate to promote successful remodeling.

This strategy greatly improved the efficacy of these stents and reduced the need for

anticoagulation or anti inflammation therapies. However, with biodegradable alloys, it

becomes increasingly important that the degradation products as well as the degraded

stent be compatible with the vascular environment. It is especially important that the

stent material be compatible chemically with the vascular environment and does not

cause significant toxicity to the endothelium or the circulating blood107–109. Polymers

have been considered as potential candidates to develop degradable stents.

Polyamides, Polyolefins, Polyesters, Polytetrafluoroethylene, Poly(L-Lactide) and

Polyurethanes have all been utilized to make vascular stents. However the unknown

long term toxicity as well as poor mechanical behavior of the polymeric materials have

deterred development of polymeric stents71,110. Therefore, there is more recent interest

in biodegradable metals for the fabrication of stents.

The first patent for a degradable metallic stent was issued in 2002. Several iron

and magnesium based metallic alloys have been investigated since then107,111. The

rationale behind the selection of these metals was their natural presence in the body

and their positive contribution to bodily functions. Iron is an essential element in the

Page 45: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

45

body and has known to cause relatively low cytotoxicity and therefore was among the

first degradable element investigated as candidate material for stents105,112. Magnesium

is also an essential element in the body with relatively low toxicity levels and it is also

efficiently excreted out in the urine. Additionally, magnesium has known to positively

impact cardiovascular mechanisms in the body113,114. Therefore, more recently

magnesium is gaining popularity as a base element for vascular stents.

In Chapter 4 of this dissertation, a novel magnesium alloy is evaluated for its

potential use as a vascular stent material. This chapter deals with assessment of

hemocompatibility of this alloy for its suitability for use in a vascular environment.

Parameters such as surface finishing conditions greatly influence the chemical

composition of the surface. For example, electropolishing of 316 L stainless steel

leaches the surface of most metallic elements except chromium. This process leads to a

formation of a layer of chromium oxide when the stent is exposed to external

environment, preventing further corrosion115. Other finishing processes such as

abrasive polishing cause an enrichment of elements on the surface as well as randomly

oriented crystals at the interface. These parameters such as the microtopography of the

surface, local chemical composition, roughness and crystal size offer important

mesoscale cues which subsequently alter cell behavior. Surface defects as well as trace

elements such as elements at grain boundaries have been known to cause alterations

in proteins and affect cellular response. Therefore the formation of an oxide layer is

beneficial in degradable metallic implants107.

Page 46: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

46

Future strategies for development of biodegradable metallic stents are focused

on structured, long-term and multidisciplinary investigation of implant efficacy in vivo.

These studies are particularly useful in clinical translation of this technology.

Macroscale Systemic Cues

The capability of cells to sense environment and produce signals that influence

cell behavior, phenotype and cell fate is one of the most important ways to modulate cell

response. The previous sections described the effect of soluble small molecule triggers

and biomaterial scaffolds on cellular responses critical for tissue engineering and

regeneration. However, cells exhibit a high level of sensitivity to biophysical cues from

the environment such as mechanical forces, physical alterations and gravitational

stresses. In the past decade, the mechanobiological cues that influence cell behavior

have been identified and used favorably to promote regeneration27,116. The major

roadblock to generation of functional tissue engineered tissues that can be used in a

clinical setting has been the limited understanding of physicochemical cues that affect

tissue development as well as the high cost of the existing tissue engineered

products25,117. Bioreactors provide reproducible, standardized, and controlled

biomechanical and environmental stimuli to cells in a closely monitored system. Some

of these environmental cues include, pH, temperature, waste removal, nutrition supply,

mechanical stresses, gravitational changes, etc. The great degree of reproducibility,

environment control and automation provided by bioreactors has led to their application

in large scale applications118–121.

Bioreactor systems historically have been employed in various applications such

as fermentation, food processing, waste water treatment, drug testing as well as protein

synthesis. They eliminate the need for in vivo testing at initial stages of development of

Page 47: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

47

therapeutics by simulating an organotypic environment118,122,123. With respect to tissue

engineering, 3D tissue engineered structures are typically cultured on porous scaffolds

which provide a support to house the cells. While initially these were initially cultured on

a static 2D culture environment, with increased interest in biomimetic designs,

bioreactor culture became more popular. Long-term bioreactor culture of these

structures promoted efficient nutrient transfer, ability to provide mechanotransducive

forces as well as close monitoring of tissue growth in an in vitro model that mimicked

the body more closely than tissue culture plates. In the human body, cells are constantly

subjected to mechanical, electrical and chemical forces that are essential for their

synchronized metabolic functions. Many types of bioreactor systems have been

developed in the past. Some of the earliest important bioreactors include, the spinner

flasks, rotating wall vessel bioreactor, hollow fiber bioreactors, direct perfusion systems

as well as custom designed bioreactor systems 122–124 as shown in Figure 2-7 below.

Each of the bioreactor systems mentioned have unique properties such as increased

turbulence as well as convective mass transfer (Spinner flasks), low shear environment

(Rotating wall vessels), precise mechanical control (Custom designed bioreactors and

Perfusion systems). The limitations as well as the advantages of bioreactors depend on

their specific applications. However, any bioreactor should follow one of the following

functions according to Freed : (1) provide a framework for uniform distribution of cells on

a three-dimensional scaffold; (2) maintain the desirable concentration of gases and

nutrients in the culture medium; (3) stimulate efficient mass transfer to the growing

tissue; (4) presenting developing tissue with controlled physical stimuli; and (5) provide

Page 48: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

48

information regarding the formation process of 3D tissues, which originate from the

isolated cells124,125.

Figure 2-7. Some representative bioreactor systems for tissue engineering. (A) Spinner flasks; (B) Rotating wall vessel bioreactors (C) Hollow-fiber bioreactor (D) Perfusion systems (E) Custom designed computer-controlled system. Image reference: Martin I, Wendt D, Heberer M. The role of bioreactors in tissue engineering. Trends Biotechnol. 2004;22(2):80-86. doi:10.1016/j.tibtech.2003.12.001.

One such bioreactor system gaining popularity as an ideal vehicle for tissue

engineering are rotating wall vessel bioreactors (RWV)119,126,127. Initially, the motive for

the fabrication of microgravity bioreactors was to create a system that can simulate the

Page 49: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

49

changes in gravitational forces in outer space on earth. This was developed by NASA

as a cost-effective model system to study and preliminarily experiments space flight

experiments. But over the recent years, space research has also contributed to

application of some of its principles in tissue engineering127. Microgravity provides a

unique environment that promotes efficient nutrient transfer, cell-cell signaling in a low

shear stress environment. In real microgravity, cells have been known to aggregate and

initiate differentiative signaling which can cause 3D tissue constructs. Further

microgravity has also known to promote stem cell proliferation which also is an

important for tissue engineering126,128–130.

In 1970, NASA’s Johnson center assembled a team of scientists to develop an

earth based system for simulating some aspects of microgravity to pretest space based

tissue engineering studies. The first two bioreactor models developed by NASA were

called Slow turning lateral vessels (STLV) and High aspect ratio vessel (HARV)119,127.

The RWV bioreactors of two concentric cylinders which rotate at a constant

angular speed. The reactor also consists of an axial silicone membrane for gas

exchange. The vessels can be fabricated in different volumes and speed of rotation is

also adjusted per the system used. Initial studies with simulated microgravity

bioreactors hypothesized that the expected damaging hydrodynamic forces on cells

would be from high velocity gradients and associated shear stress involved in the

bioreactors. The appearance of this shear stress is through the boundary layer that

exists between fluid moving inside the vessel relative to the vessel wall. The RWV

bioreactor however eliminates this gradient resulting in a reduced shear stress

environment131,132.

Page 50: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

50

Figure 2-8. Path of a particle or cell aggregate in a RWV bioreactor illustrating the net forces acting on the particle in rotation.

The diagram above represents the path of a particle or cell aggregate and the net

forces acting on the particle in rotation is also illustrated. The sedimentation velocity due

to gravity, Vs, is composed of an inwardly directed component, Vsr, and a tangential

component, Vst. There is an outwardly directed motion, Vcenr, produced by centrifugal

force, and a tangential component, Vcort, from the Coriolis force. Coriolis force is an

inertial force (also called a fictitious force) that acts on objects that are in motion relative

to a rotating reference frame the resolution of these forces on cells or aggregates as

shown produces a slow descent through the culture media as the vessel turns at a pre-

determined speed.

Because the net forces on the cells are substantially reduced, this culture

environment is sometimes referred to as simulated or modeled microgravity. The results

obtained using simulated microgravity conditions utilizing the RWV have been

confirmed and validated by experimental models in space flights to outer space133–136.

Page 51: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

51

The initial attempts to engineer tissues in simulated microgravity, led to culturing

of cartilaginous tissue constructs consisting of a collagen and glycosaminoglycan base

in 1997126,137. This study showed positive results with higher regenerative tissue

formation compared to other methods. Interestingly, the study showed that the cells

formed 3D aggregates as the cells were cultured for longer times in microgravity. This

system has also been utilized to culture stem cells and vascular cells for vascular tissue

engineering. In a recent study by Grimm et al., EA.hy926 endothelial cell lines cultured

under simulated microgravity formed tube-like structures with walls mostly consisting of

single-cell layers. They were found to have a lumen and resembled the intima of blood

vessels in vivo, reaching a length of up to several centimeter138,139. More interestingly, a

recent study by Chiu et al., showed that human cord blood stem cells when cultured

under simulated microgravity proliferated and trans-differentiated into 3D structures

resembling vascular tubules while exhibiting vascular endothelial phenotype

expressions140. These studies have opened exciting avenues for the use of simulated

microgravity bioreactors as a controlled environment for vascular tissue engineering.

Additionally, this environment has also known to support bone tissue growth,

which was studied by culturing osteoblasts cell in the RWV reactor141. These systems

have also gained popularity in developing cancer models due to the improved mass

transfer characteristics and 3D growth environment129,142. Many excellent recent reviews

have summarized the effect of simulated microgravity on the behavior of various cell

systems128,129,143,144. In Chapters 6 and 7 of this dissertation, the effect of simulated

microgravity on blood derived endothelial cell systems are investigated and reported.

The goal of this study was to use simulated microgravity as a model system to

Page 52: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

52

understand the behavior of these cells to further study the vascular repair mechanisms

enabled by these cells. This study has the potential to develop regenerative therapy for

tissue engineering.

Future studies using microgravity mediated tissue engineering rely on confirming

the efficacy of these constructs in enabling vascular repair in patients. Current studies in

this field is focused on translating these strategies to production of whole sensory

organs and other high-density structures.

Page 53: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

53

CHAPTER 3 NANOSCALE LOCAL CUES- DEVELOPMENT OF A NOVEL VEGFR2 AGONIST

APTAMER ASSEMBLY

Introduction

In addition to intrinsic factors produced within cells that regulate the normal

functioning of cell systems, living cells are highly sensitive to extrinsic cues presented to

them at the molecular scale by the surrounding environment10,17,18. The interactions of

these nanoscale triggers with cellular systems are highly efficient in eliciting a multiscale

and complex response within the body. These triggers can range from biochemical cues

such as soluble growth factors, cytokines and other small molecules to nano-structural

cues provided by the ECM. Due to the small scale of these interactions, there is still a

dearth of information in understanding these mechanisms in vivo10,14,18. However,

recent advances in tissue engineering, regenerative medicine, and cell based therapies

necessitate the development of novel strategies based on delivery of these nanoscale

cues that allow for controlling cellular processes for an application of interest. The end

goal is to design approaches that are targeted, specific, robust, and ultimately result in

molecular pathway activation events and the transduction of signals that direct cellular

responses, such as cell proliferation, differentiation, tissue matrix production or even cell

death. For receptor-mediated cell processes, the identification of stable receptor

substrate ligands that exhibit precise binding, high specificity, and targeted activation of

a particular receptor represents an area of active research145. Current efforts in this

field, aim to use receptor-specific growth factors and cytokines or their respective

receptor-binding peptide sequences, as these are responsible for native interactions

that result in receptor activation. While inherently successful as receptor agonists,

these agents, often being difficult to control spatially, can lead to off-target effects

Page 54: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

54

through the activation of nonspecific receptors and binding to non-specific cell

types34,146. Unfortunately, identification of therapeutic agents that selectively activate

specific cell surface receptor targets has proven to be quite a challenge. Overcoming

these limitations in the control of receptor-mediated processes will result in distinct

improvements in directing cellular processes such as proliferation, migration, and

differentiation.

A promising approach to achieving this goal that is gaining in popularity in the

recent times is the use of nucleic acid aptamers. Aptamers are short single-stranded

oligonucleotides that exhibit highly specific binding to intracellular and extracellular

target sites147. The high affinity and specificity of aptamer binding can be attributed to

their ability to form complex three dimensional structures51. This is typically due to their

capability to form complementary base pairs allowing them to form a variety of

secondary structures that can be further formed into three dimensional assemblies.

These assemblies are recognized by their cognate targets and form complexes, similar

to that formed when antibodies interact with antigens. The aptamer-target interaction

can be via hydrophobic and electrostatic interactions, hydrogen bonding, van der Waals

forces, shape complementarity and base stacking. Such interactions enable the specific

binding of the aptamer to the target with high affinity.

Ever since their isolation using the SELEX (Systematic Evolution of Ligands by

Exponential Enrichment) process, which was developed in the lab of Larry Gold at the

University of Colorado148, aptamers have been studied extensively as a diagnostic and

therapeutic tool for a wide range of applications35,53,149. In addition, using this method,

aptamers have been selected against a wide array of targets from small molecules,

Page 55: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

55

proteins and bacteria, to whole cells (i.e. whole-cell SELEX)36,47,150. The major

advantages in using aptamers over traditional antibodies and growth factors are their

high affinity and targeting specificity as well as their non-immunogenic nature. Aptamers

can target and bind to small hidden domains within a large biomolecule that can be

inaccessible to larger antibodies. Aptamers are also easily synthesized and can be

modified to confer nuclease resistance, and thus stability to withstand filtration by the

kidney and improves circulation time in vivo47. With regard to aptamer-receptor

interactions, most work with aptamers exploits the ability of aptamers to inhibit protein-

protein interactions, such as receptor-ligand interactions, thereby rendering them

receptor antagonists47. This approach has been quite successful in the area of

neovascularization and angiogenesis inhibition151,152. Pegaptanib (Macugen) targeted

against VEGF, is currently the only federally approved aptamer therapeutic drug153. The

higher affinity of aptamers to their targets as compared to other receptor ligands, such

as antibodies or growth factors, makes them excellent at blocking receptor activation.

However, the therapeutic ability of agonist or receptor- activating aptamers is yet to be

explored completely.

As a departure from current strategies, this work shows the ability of an

engineered aptamer assembly to act as a receptor agonist that activates VEGFR2 on

human endothelial cells. The activation of VEGFRs by VEGF plays a crucial role in

many processes of vascular stem and mature cells, including cell proliferation,

migration, differentiation, survival, tissue factor production, and nitric oxide production,

as well as functions such as angiogenesis39. The delivery of VEGF, local as well as

systemic, has been extensively investigated in order to improve angiogenesis for

Page 56: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

56

neovascularization in vascular tissue engineering. Although the strategy has proven to

be highly effective in vitro, the clinical translation still has some gaps yet to be filled. For

example, in a study by Simón-Yarza et al. (2012)37, VEGF delivery for clinical trials for

myocardial regeneration revealed no significant improvement with respect to placebo.

Intravenous administration of the growth factor also proved to be ineffective due to its

short in vivo half-life (~30 min) and overall dose was limited by off-target site toxicity

issues. Therefore, there in a need to identify alternate strategies to specifically cause

receptor activation to improve angiogenesis and other downstream functional

responses. With that in mind, the following aim was established and tested.

Specific Aim 1

Design and develop of a novel divalent DNA oligonucleotide sequence (aptamer),

that when assembled into a unique divalent assembly acts as a receptor agonist towards

VEGFR2.

Materials and Methods

Aptamer Design

The aptamers used in this study include an 80 mer VEGFR2 binding DNA

aptamer with modified nucleic acids that was obtained from Aptamer Sciences (AptSci,

Pohang, Gyeongbuk, South Korea) and was used to confirm endothelial cell binding.

The sequence (Apt80mer) is shown in Table 3-1. Following validation of endothelial cell

binding with the commercially available aptamer sequence, subsequent receptor

agonist studies were conducted with a modified 80mer DNA aptamer of similar

sequence, however replacing the benzyl containing modified bases (BndU) (indicated

as 5 in the sequence) with thymine (Integrated DNA Technologies, Coralville, Iowa).

The modification of the bases results in slightly lower binding affinity, however the

Page 57: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

57

binding specificity towards VEGFR2 is maintained. The modified sequence (AptM80mer) is

also shown in Table 3-1.

Table 3-1. Aptamer sequences and configuration

Towards the goal of assessing aptamer agonist function, aptamer assembly

composed of two identical AptM80mer monomers tethered to each end of an 18-atom

hexa-ethylene glycol spacer thus creating a divalent assembly (AptDivalent) (Integrated

DNA Technologies) was fabricated. Polyethylene glycol (PEG) was chosen to be the

spacer of the divalent aptamer assembly because it exhibits many qualities desirable for

the functionality of the assembly. First, PEG shows favorable biological properties in

that it is non-toxic and non-immunogenic. Additionally, the hydrophilic nature of PEG

helps to support water solubility of the assembly. Further, PEG is flexible and does not

impose steric hindrance on aptamer molecules bound to it. Finally, PEG molecules

have been shown extensively in the literature to prevent non-specific binding and thus

may prevent non-specific aptamer binding154. As another selection criterion, the root

mean square distance between the two subunits of VEGFR2 is reported as 3.5 Å155.

Therefore, to make possible the binding of both receptor subunits, the 18-atom hexa-

Aptamer Description Sequence

Apt80mer 80mer monomer 5’ GAT GTG AGT GTG TGA CGA GC5 ACG ACG 5C5 GG5 G5A A55 5A5 AAA GAC AC5 G5G 5A5 A5C AAC AAC AGA ACA AGG AAA GG 3’

AptM80mer Modified 80mer monomer

5’ GAT GTG AGT GTG TGA CGA GCT ACG ACG TCT GGT GTA ATT TAT AAA GAC ACT GTG TAT ATC AAC AAC AGA ACA AGG AAA GG 3’

AptDivalent 5’-3’/spacer/5’-3’ 5’ GAT GTG AGT GTG TGA CGA GCT ACG ACG TCT GGT GTA ATT TAT AAA GAC ACT GTG TAT ATC AAC AAC AGA ACA AGG AAA GG 3’/ (PEG)6 spacer /5’ GAT GTG AGT GTG TGA CGA GCT ACG ACG TCT GGT GTA ATT TAT AAA GAC ACT GTG TAT ATC AAC AAC AGA ACA AGG AAA GG 3’

Page 58: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

58

ethylene glycol spacer with an end-to-end chain length of 21 Å and a calculated Flory

radius of 10.26 Å was selected, as this length would make such a dimerization

mechanism possible156. It should be noted that while this mechanism is not

demonstrated in this work, based upon the length of the spacer, it is possible with the

AptDivalent assembly. The predicted secondary structure with the lowest free energy, and

a schematic representation of the divalent aptamer assembly are shown in Figure 3-1 A

& B.

Figure 3-1. Aptamer structure. (A) The predicted secondary structure with the lowest free energy of the AptM80mer monomer (structure predicted by UNAfold) (B) Schematic representation of the divalent aptamer assembly (AptDivalent) showing two AptM80mer monomers tethered through the 3’ and 5’ ends to an 18-atom hexa-ethylene glycol spacer.

Aptamer-Endothelial Cell Binding

To determine the ability of the 80mer aptamer and the modified base 80mer,

Apt80mer and AptM80mer, respectively, to bind to the VEGFR2 receptor, flow cytometric

analysis was performed using human umbilical vein endothelial cells (HUVEC) (Lonza,

Baltimore, MD). HUVEC between passages 5-9 were cultured in endothelial cell basal

media (EBM-2) (Lonza), supplemented with hEGF-2, hydrocortisone, hFGF-b, VEGF,

IGF-1, ascorbic acid, and gentamycin/amphotericin-B, and 2% fetal bovine serum (FBS)

in 37°C CO2 incubator. The VEGFR2 binding aptamer (Apt80mer) and the modified base

aptamer (AptM80mer) containing a terminal amine group were obtained to allow for

A B

Page 59: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

59

fluorescein isothiocyanate (FITC) modification. The FITC tag was incubated with the

amine-terminated aptamers separately in borate buffer (pH 8.5) for 1 h at room

temperature with gentle mixing. The excess FITC was removed via the use of three

consecutive Zeba desalting columns (Pierce biotechnology, Rockford, IL) to ensure

complete removal of the dye. The amine group on the aptamer was then linked to the

isothiocyanate group forming a thiourea type bond between the FITC molecule and the

aptamer. The FITC conjugated aptamer solution was suspended in phosphate buffered

saline (PBS) (pH 7.4) and stored in the dark. Control experiments included the use of

VEGFR2 antibody (CD309) and an IgG isotype control antibody for testing non-specific

binding (BD Biosciences, San Jose, CA). In preparation for flow cytometry, HUVEC

were detached from tissue culture polystyrene (TCPS) flasks using cell dissociation

media, then split into separate micro-centrifuge tubes, each containing a suspension of

300,000 cells. The FITC conjugated aptamers i.e.; Apt80mer and AptM80mer previously

prepared were then incubated with HUVEC for 1 h the in suspension to allow for the

aptamer binding to the cell surface VEGFR2. This was followed by a washing step

(three centrifugal spins and washes) utilizing PBS containing 0.1 % Tween-20 to ensure

the removal of non-specifically bound aptamer. The cells were suspended in PBS

containing 4% FBS and run on an Eclipse EC800 flow cytometer (Sony Biotechnology,

Champaign, IL). The data was analyzed using FlowJo (Treestar, Ashland, OR) and

plotted as a histogram. To confirm binding specificity of the Apt80mer and AptM80mer, in a

separate experiment sample, HUVEC were treated as described, except the cells were

incubated with 2 ng/mL soluble VEGF prior to incubation with the aptamer to block the

target VEGFR2 receptor. In another experiment, AptM80mer binding when in competition

Page 60: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

60

with soluble VEGF was assessed. In this experiment, HUVEC were incubated

simultaneously with 150nM AptM80mer and 50, 75, 100 or 150nM soluble VEGF. Flow

cytometry was conducted to detect changes in aptamer binding affinity in the presence

of soluble VEGF and the data was analyzed using FlowJo.

VEGFR2 Phosphorylation

The activation of VEGFR2 by both the monomeric form and divalent aptamer

assembly was tested by assessing phosphorylation events of VEGFR2 on HUVEC.

Phospho-VEGFR2 was quantitatively measured spectrophotometrically using Human

Phospho-VEGF R2/KDR DuoSet IC (R&D Systems, Minneapolis, MN). Briefly, HUVEC

were grown in complete media previously described to nearly 100% confluence on

TCPS. The cells were then serum starved overnight in basal media containing 0.2%

FBS and 0.1 % Bovine serum albumin (BSA) (Sigma Aldrich, St. Louis, MO).

Immediately prior to stimulation, the HUVEC were washed with PBS, and then

stimulated for 10 min in either unsupplemented basal media (BM), unsupplemented

basal media containing 100 ng/mL VEGF, unsupplemented basal media containing 300

nM AptM80mer or unsupplemented basal media containing 150 nM AptDivalent. Following

stimulation, the cells were first rinsed with ice cold PBS, and then incubated on ice for

15 minutes in ice cold lysis buffer containing 10 μg/mL each of protease inhibitors

Aprotinin and Leupeptin (R&D Systems). The lysates were collected and assayed by

following the standard ELISA protocol provided by the manufacturer (R&D Systems).

The optical density of the samples was immediately determined using a Synergy H1

microplate reader (Biotek) with the absorbance measured at 450 nm and wavelength

correction set at 540 nm.

Page 61: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

61

Signal Pathway Activation

Activation of the downstream Akt pathway by the divalent aptamer assembly was

assessed via immunological labeling and flow cytometry. HUVEC were grown to 80%

confluence on TCPS prior to stimulation, and then subsequently, serum starved

overnight in EBM-2 devoid of FBS and growth factors. Following culture overnight in

serum free media, the cells were stimulated for 20 min in either unsupplemented serum

free basal media (EBM-2), unsupplemented basal media containing 2 ng/mL VEGF, or

in unsupplemented basal media containing 300 nM AptDivalent. Following the stimulation,

the cells were washed with PBS, trypsinized and split into microcentrifuge tubes

containing 300,000 cells. The stimulated cells were permeabilized using a cold 100%

methanol solution at 4°C for 15 minutes, washed 3 times to remove excess methanol

and fixed using fluorofix buffer (Sony Biotechnology, Champaign, IL) for 30 minutes at

room temperature. The cells were again washed using PBS three times to remove any

excess fixative in preparation for staining. The cells were then probed using Anti-

phospho Akt antibody (Ser473) (EMD Millipore, Billerica, MA) (250 µg/sample) for 1 h in

the dark with occasional gentle mixing. The cells were subsequently washed 3 times

with PBS containing 0.05 % Tween-20 to remove non-specifically bound antibody. The

cells re-suspended in PBS containing 4% FBS, were subsequently run on the Eclipse

EC800 flow cytometer and the data was examined and plotted using FlowJo.

Endothelial Nitric Oxide Synthase Expression

Changes in expression of endothelial nitric oxide synthase (eNOS) were

assessed using western blot analysis. Briefly, HUVEC were grown in complete media

previously described, to 80% confluence. Prior to stimulation, the HUVEC were washed

with PBS, and then stimulated for 24 h in either unsupplemented basal media (EBM-2

Page 62: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

62

devoid of serum and growth factors), unsupplemented basal media containing 2 ng/mL

VEGF, or in unsupplemented basal media containing 300 nM AptDivalent. Following

stimulation, cells were lysed on ice for 15 min using 0.1% triton-X in PBS, containing

10μg/mL protease inhibitor cocktail (Sigma Aldrich). Cellular proteins were resolved by

7.5% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and subsequently

transferred to a polyvinylidene difluoride (PVDF) membrane (Fisher Scientific, Waltham,

MA). After transfer, the membranes were divided with the top portion being

immunoblotted with anti-eNOS primary antibody (1:500, Abcam, Cambridge, UK)

overnight at 4°C. The lower portion of the membrane was probed with anti β-tubulin

primary antibody to serve as a loading control (1:500, Abcam). eNOS and β-tubulin

protein expression was visualized using a horseradish peroxidase–conjugated goat anti-

rabbit secondary antibody (1:5000, Abcam) and colorimetrically detected using the Opti-

4CN HRP substrate detection kit (Bio-Rad, Hercules, CA). The membrane was digitally

imaged and the intensity of the resulting bands was quantified using Image J software

Version 1.46.

Capillary Tube Formation (Angiogenesis)

To assess the function of the divalent aptamer assembly, a capillary tube

formation assay was conducted. Recall that, the divalent aptamer is composed of two

AptM80mer tethered at both ends of a PEG spacer. Therefore, to confirm the effectiveness

of the divalent assembly structure over the monomeric aptamer, the AptM80mer was also

included. Briefly, HUVEC were plated on a 48-well tissue culture plate pre-coated with

250 µl of Cultrex basement membrane extract (BME) (Trevigen, Gaithersburg, MD). The

cells were then treated with 100 µl each of either unsupplemented basal media (EBM-2

devoid of serum and growth factors), unsupplemented basal media containing 2 ng/mL

Page 63: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

63

VEGF, unsupplemented basal media containing 300 nM AptM80mer or unsupplemented

basal media containing 150 nM AptDivalent. After 12 h incubation in a 5% CO2 humidified

atmosphere at 37°C, the three-dimensional structures formed by the cells in this matrix

were examined using an inverted phase-contrast microscope and digital images were

recorded. Tube-like structures were further analyzed by measuring the sum of the

lengths all tubules per field using the image analysis software AxioVision, (Zeiss, V4.5)

(Carl Zeiss Microsystems, Thornwood, NY). Four randomly selected low-power fields

were examined for each sample.

Chorioallantoic Membrane (CAM) Assay

Fertilized chicken eggs (30 pieces) were obtained from a local farm (Laughing

Chicken Farm, Gainesville, FL) and placed in trays in a horizontal position in an

incubator maintained at 37 °C with approximately 60–65 % humidity after cleaning with

70 % ethanol. The eggs were incubated for 3 days in horizontal position and were

rotated along their axis twice per day. On day three of incubation, 2 mL of albumen was

aspirated using a syringe needle (25 G) from the transverse end of the egg, to detach

the developing CAM attached to top part of the shell of the egg. The top part of the shell

was also gently poked with the syringe to further help the dropping of the CAM. Then, a

small window of around 1.5 cm2 was gently opened on the top shell with sterile forceps

on the blunt end of the egg without damaging the embryo. Whatman Type I filter paper

was used as carriers for testing angiogenic response. The filter was punched using a ¼

‘paper punch. The paper discs were gas sterilized using EtOH gas and soaked in 100 µl

each of either normal saline solution, saline solution containing 2 ng/mL VEGF, saline

containing 150 nM AptDivalent. The discs were then placed directly on top of the CAM.

The window was then sealed with transparent sterile band-aid to prevent dehydration

Page 64: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

64

and possible infections and returned to the incubator. The eggs were incubated for

another 3 days, the covering band-aids were then removed, and the filter paper was

then gently removed. The surrounding vessels of the CAM were then examined under

the dissecting microscope and recorded (Leica M320). The images were then

qualitatively analyzed to assess angiogenic response of the aptamer in comparison to

the controls.

Results and Discussion

Herein the design of a supramolecular aptamer assembly for targeted VEGFR2

binding and activation has been reported. The design of the aptamer assembly is based

upon the native interaction of soluble VEGF with both subunits of surface bound

VEGFR2. When bound, soluble VEGF acts as a bridge between the two subunits and

results in their dimerization. The rationale behind this approach has proven successful

in other surface receptors, such as OX40, a receptor expressed on activated T-cells61.

In this work, an RNA aptamer scaffold was designed with agonist function by binding to

the receptor subunits of the OX40 receptor. In a similar manner, it is hypothesized that

the aptamer assembly described herein allows for the VEGFR2 binding aptamer at each

end of the 18 atom spacer to bind to each of the 2 subunits comprising the VEGFR2

and facilitate dimerization. A schematic demonstrating the proposed mode of action of

the divalent aptamer assembly is shown in Figure 3-2. Receptor agonist activity is

predicated on the ability of the aptamer assembly to first bind the target receptor.

Page 65: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

65

Figure 3-2. Schematic representation of the proposed method of action of the divalent aptamer assembly. Binding to target and activation via dimerization, membrane bound VEGFR2.

The Apt80mer used in this work has a high VEGFR2 binding affinity with a Kd of

0.12 nM. (AptSci) However the divalent aptamer assembly is composed of two identical

monomeric units of AptM80mer tethered to each end of an 18-atom hexa-ethylene glycol

spacer. To demonstrate specificity of the Apt80mer as well as the AptM80mer monomer

towards HUVEC, known for high levels of VEGFR2 expression, flow cytometry was

performed on HUVEC probed with FITC conjugated Apt80mer (FITC-Apt80mer) and FITC

conjugated AptM80mer (FITC-AptM80mer). Controls included phycoerythrin (PE) labeled

VEGFR2 specific antibody and PE-labeled isotype control. The results from the study

are shown below in Figure 3-3. The images below show representative histograms from

flow cytometric analysis. The results confirm VEGFR2 expression on HUVEC as

evidenced by the rightward shift (~82% positive) for the VEGFR2 antibody, compared to

~6% positive for the isotype control (Figure 3-3 A and B).

Page 66: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

66

Figure 3-3. Aptamer-cell binding interactions. Images A-F show flow cytometry data of aptamer-endothelial cell binding. Un-probed HUVEC are shown in each panel as the blue histogram. The red histogram shows (A) positive binding of CD309 (VEGFR2) antibody; (B) negative binding from the isotype control; (C) positive binding of FITC conjugated Apt80mer aptamer; and (D) inhibition or decreased binding of the aptamer when blocked with soluble VEGF prior to aptamer incubation. (E) positive binding of FITC conjugated modified AptM80mer aptamer (F) inhibition or decreased binding of the aptamer when blocked with soluble VEGF prior to aptamer incubation. Images G-J show flow cytometry data of aptamer-endothelial cell binding in competition with soluble VEGF. Un-probed HUVEC are shown in each panel as the blue histogram. The red histogram in each figure shows positive binding of 150 nM of the fluorescent tagged AptM80mer solution in competition with (G) 50 nM (H) 75 nM (I) 100 nM (J) 150 nM VEGF solutions.

PE -18.0

A

PE +

82.0 PE + 6.59

B

PE -93.4

FITC - 10.6

C

FITC+ 89.4

FITC - 79.5

D

FITC+ 20.5

FITC – 37.5

E

FITC + 62.5

FITC - 84.2

FITC + 15.8

F

G

I

H

J

Page 67: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

67

To demonstrate binding specificity of the Apt80mer and AptM80mer towards the

VEGFR2 target receptor, HUVEC were incubated with Apt80mer and AptM80mer with and

without pre-treatment with soluble VEGF. The flow cytometry data demonstrate ~89%

positive targeted binding of Apt80mer to VEGFR2 compared to the diminished ~20%

positive Apt80mer binding when pre-treated with soluble VEGF (Figure 3-3 C and D).

Similarly, the data also showed ~62.5% positive targeted binding of AptM80mer to

VEGFR2 while the cells pre-treated with soluble VEGF showed diminished binding

(~15.8% positive) (Figure 3-3 E and F). This data demonstrates successful receptor-

ligand binding, which is the critical first step in the complex process of membrane

receptor activation157. In addition, the cells pre-incubated with VEGF showed diminished

aptamer binding, which indicates specificity of the binding aptamer to VEGF receptor.

Finally, the assessment of competitive binding of the aptamer in the presence of

VEGF at varying concentrations showed that VEGF did not interfere with the aptamer

binding at concentrations ranging from 50 to 150 nM (Figure 3-3 G-J). Also, preliminary

data from a separate experiment conducted, indicated that over a period of 20 h, the

bound monomeric aptamer (AptM80mer) (FITC tagged) is internalized by the cell. This

early finding is consistent with reports in the literature that showed internalization of

aptamers by the target cell50,57. Further investigation is required to study the specific

localization of the aptamer assembly within a cell.

Page 68: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

68

Figure 3-4. Aptamer Induce VEGFR2 activation and downstream molecular events (A) Quantification of HUVEC VEGFR2 phosphorylation upon exposure to either unsupplemented basal media (BM), or BM supplemented with 100 ng/ml soluble VEGF, 150 nM soluble divalent Aptamer (AptDivalent) or 300 nM Monomer (AptM80mer). Data shown are Mean ± SD; Images B and C show activation of Akt signal transduction pathway. HUVEC are shown in each panel as the blue histogram. The red histogram shows positive binding of Anti-Phospho Akt (Ser473) antibody, thus indicating pathway activation upon stimulation with (B) 2 ng/ml soluble VEGF; and (C) 300 nM AptDivalent. Image D represents eNOS protein expression from HUVEC treated with either unstimulated basal media (BM), or BM supplemented with 2 ng/ml soluble VEGF, or 300nM soluble divalent Aptamer (AptDivalent). The top membrane shows eNOS specific protein bands and the bottom membrane shows beta tubulin specific bands that were used as a loading control. eNOS protein band

intensity was normalized to β-tubulin and quantified using image J software

version 1.46. In figures A and D, based upon One-way ANOVA with Tukey’s multiple comparison test, N.S. indicates no significant difference (p>0.05), and * asterisks indicate a statistically significant increase (p<0.05).

The binding of soluble VEGF to VEGFR2, initiates a sequence of events resulting

in receptor dimerization, kinase activation, and auto-phosphorylation of specific tyrosine

kinase residues within the dimeric complex158. The activation of tyrosine kinase through

C D

Page 69: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

69

trans-phosphorylation between receptor molecules is the first step leading to activation

of intracellular signal transduction pathways157. Therefore, to assess the activation of a

cell surface receptor, assessment of phosphorylation events is important. Towards this

goal HUVEC were treated with basal media supplemented with either the AptDivalent,

AptM80mer, or soluble VEGF and assessed for VEGFR2 receptor phosphorylation. As

expected, treatment of HUVEC with soluble VEGF results in activation of VEGF

receptor-2 phosphorylation events (Figure 3-5 A).

The treatment of HUVEC with AptDivalent also results in phosphorylation of

VEGFR2 (Figure 3-4 A). Further, treatment of HUVEC with the divalent assembly

results in significantly greater phosphorylation relative to the amount of phosphorylation

from basal media or basal media containing monomeric AptM80mer (Figure 3-4 A,

p<0.05). These data reaffirm the specificity of the divalent aptamer assembly and

demonstrating the promotion of VEGFR2 phosphorylation events on HUVEC.

More importantly these data demonstrate the unique advanced ability of the

divalent assembly in receptor activation that is not seen in the monomeric form. In fact,

despite its ability to bind VEGFR2, the addition of the monomeric aptamer (AptM80mer),

from which the divalent assembly is composed, resulted in no difference in

phosphorylation over unsupplemented basal media alone (Figure 3-4 A; p>0.05). The

induction of receptor phosphorylation via an aptamer assemblage shown here

represents one of the early and few examples in the literature of receptor activation via

an aptamer complex.

The downstream events in VEGF receptor activation are quite complex, involving

multiple signal molecules and multiple molecular pathways, including the well

Page 70: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

70

characterized Akt pathway39. To further assess the effect of aptamer-receptor binding,

the activation of this downstream molecular pathway was examined. Akt pathway

activation is involved in numerous diverse biological processes, such as cell

survival/apoptosis, cell cycle control, angiogenesis, differentiation, and cell growth and

proliferation159,160. In addition, Akt pathway activation has been associated with

activation of VEGFR2, through treatment with soluble VEGF161,162. In this study, serum

starved HUVEC were treated with AptDivalent or soluble VEGF, then phosphorylation of

Serine 473 is detected with a phosphor-Akt antibody and analyzed via flow cytometry.

Data reveal that treatment with the AptDivalent results in activation of the Akt pathway in

levels similar to that seen with soluble VEGF (Figure 3-4 B and C). The cells treated

with either AptDivalent or soluble VEGF showed positive staining for phosphor Akt, ~97%

and 93% respectively. Although the activation of Akt pathway by soluble VEGF is

expected and has been shown by others, there are currently no reports of Akt pathway

activation triggered by treatment with a DNA aptamer, nor an aptamer assembly. These

promising results provide early evidence that the AptDivalent assembly as designed

promotes activation of VEGFR2 and causes downstream activation of pathways that

control critical cell functions.

eNOS is an important protein involved in the production of Nitric oxide in

endothelial cells, and is critical for regulation of cardiac function and angiogenesis. The

protein expression experiments reveal aptamer induced upregulation of eNOS to levels

comparable with what is seen in VEGF induced upregulation (Figure 3-4 D).

Quantification of the protein levels shows a statistically relevant increase in eNOS

expression in HUVEC stimulated with VEGF or the divalent aptamer assembly. These

Page 71: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

71

results are encouraging and indicate a potential angiogenic downstream effect of

receptor activation.

As a final functional assessment of the agonist behavior of the AptDivalent, a tube

formation assay was utilized. The tube formation assay is well characterized and

regarded as a powerful tool to screen for various factors that promote or inhibit in vitro

angiogenesis. In addition, angiogenesis as measured in the tube formation assay, is

mediated by VEGF signaling through activation of VEGFR2163. The soluble AptDivalent

promotes HUVEC migration and capillary like tube formation (Figure 3-5 A-D). When

compared to HUVEC stimulated with soluble VEGF, cells stimulated with AptDivalent

shows statistically similar tube length measurements (Figure 3-5 B-C; p>0.05). The data

also indicates that the AptDivalent is more effective in induc ing capillary tube formation

than the AptM80mer (Figure 3-5 E; p<0.05). These data are particularly impactful as this is

the first report of a divalent aptamer assembly activating cellular processes involved in

promoting angiogenesis. In addition, this data also show the potency of the divalent

assembly to act as a receptor agonist while the monomeric form (AptM80mer) shows very

little or no receptor phosphorylation and downstream angiogenic effect, despite

exhibiting excellent binding affinity towards VEGFR2. These data reaffirm the rationale

behind the design of the divalent aptamer assembly. Although many robust in vitro

assays have been developed to assess angiogenic potential of various biopolymers as

well as natural substances, the process is so dynamic that it is best characterized by in

vivo studies. Although, animal studies using a mammalian subject is the golden

standard for such experiments, these are often very expensive and time consuming.

Page 72: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

72

Figure 3-5. Functional assessment of VEGFR2 aptamer binding. Representative phase contrast images of HUVEC cultured on matrigel treated with either (A) unsupplemented basal media (BM); (B) BM supplemented with 2 ng/ml soluble VEGF; (C) 150 nM soluble divalent Aptamer (AptDivalent); or (D) 300 nM Monomer (AptM80mer). Images in each panel show tube like structures formed. Quantification of tube length in µm was taken from 4 separate low powered magnification images using AxioVision, v.4.5 software. Data represent the Mean ± SD of between 18-75 measurements taken from each of 4 images. Based upon One-way ANOVA with Tukey’s multiple comparison test, N.S. indicates no significant difference (p>0.05), and * asterisks indicates a statistically significant increase (p<0.05).

The chorio allantoic membrane assay (CAM) assay ever since its development

has been widely used as an in vivo model to study angiogenesis. It has many

advantages over typical mammalian systems including low cost, and ease of

experimentation, as well as the absence of a mature immune system164,165. In this study,

C D

E

A B

Page 73: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

73

the CAM assay was used to determine the angiogenic potential of the divalent aptamer

assembly in comparison with VEGF. Saline was used as a control in this study as a

blank control. Preliminary data as shown through digital images in Figure 3-6 of the

CAM after 3 days of incubation with samples and controls indicated that the aptamer

assembly is able to induce angiogenic response comparable to VEGF.

Figure 3-6. Chorio allantoic membrane assay. Digital images of the CAM after 3 days of incubation with samples. The dotted circle marks the position of sample loaded filter paper. The arrows show neoangiogenic sprouting of vessels in the developing CAM.

The response was assessed via appearance of neovasculature around the area

where the sample was placed as shown in the images.The data presented here

demonstrate the ability of an aptamer assembly to act as a receptor agonist for

VEGFR2 on human endothelial cells. Further, the function of the aptamer to promote

endothelial cell capillary like tube formation, an important criterion demonstrating

functional agonist activity was shown. As an extension of this behavior, the CAM assay

also showed preliminary evidence of the aptamer assembly acting as a pro-angiogenic

factor in an in vivo model165. This mechanism of utilizing an aptamer agonist to

modulate receptor response and activate downstream signaling pathways in endothelial

cells has not been shown previously in the literature.

Page 74: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

74

Summary

Controlling and modulating receptor mediated response in cells is of paramount

importance in many areas such as tissue engineering, regenerative medicine,

diagnostics, etc30,33. Due to the small subcellular scale of the interactions involving

receptor mediated processes, strategies to invoke favorable cellular response via

cellular receptors are often developed using biomolecules at the nanoscale (<100 nm).

Traditional strategies for such processes historically have involved the use of cytokines

and growth factors since these represent the natural mechanism of receptor activation

in the body31,45. However, these approaches have been shown to cause nonspecific

response when used as a therapeutic means. Therefore, it is imperative to identify and

develop strategies that are targeted and specific for controlling receptor mediated cell

processes. The present work hypothesizes the use of a divalent aptamer assembly for

activation of VEGFR2 receptor in endothelial cells. This work can further be extended to

develop aptamer based receptor agonists to control other cell mediated processes such

as differentiation, proliferation, apoptosis etc. This novel strategy also has tremendous

potential for the development of novel pharmaceuticals in the treatment of a wide range

of diseases, including inflammation, autoimmune diseases, and vascular disease.

Activation of the VEGFR2 receptor has been shown to facilitate various processes such

as proliferation, migration, neoangiogenesis and differentiation in endothelial cells and

various stem cell populations. These positive outcomes of the activation event have

been exploited in designing pro-angiogenic therapies via targeted activation of VEGFR2

for vascular tissue engineering to cause repair and regeneration of vascular

tissue155,163,166.

Page 75: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

75

The results presented in this study show that this novel strategy of using nucleic

aptamers based divalent assembly to target VEGFR2 was effective in triggering the

activation cascade in these receptors in endothelial cells. The assembly was also able

to cause positive downstream angiogenic response which proves that this is a

promising strategy to modulate cell behavior favorably for vascular tissue engineering

and regenerative medicine, where controlling cell processes is of paramount

importance. The ability to modulate a functional response such as cell growth,

differentiation and angiogenesis is critical for success in these areas37,166. Therefore,

this novel, specific and targeted strategy has tremendous potential towards designing

novel nanoscale biomaterial cues for vascular tissue engineering.

Page 76: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

76

CHAPTER 4 MESOSCALE STRUCTURAL CUES- PART 1- DEVELOPMENT OF A BIOMIMETIC,

BILAYERED VASCULAR SCAFFOLD FOR TISSUE ENGINEERING SMALL DIAMETER BLOOD VESSELS

Introduction

In Chapter 3, the response of sub-cellular and molecular cues was addressed.

This chapter describes strategies to modulate cellular response by designing

appropriate cues on the mesoscale. Important cellular responses and characteristics

including alteration of cell shape, alignment, differentiation, growth and tissue formation

have been known to be modulated by architectural cues that are presented to the cell

systems by biomaterials that are in contact with them14,30,167. These structural stimuli

may be provided to them across multiple length scales. Each stimulus can be designed

to elicit a specific response from the cell system. This concept is made use of while

designing biomaterials for tissue engineering.

The biological mesoscale range has been described to be inclusive of biological

structures ranging from 10 to several 100 microns in size. Although there have been

some discrepancies in the numerical definition of the scale size itself, mesoscale is

broadly used to describe phenomenon occurring in cellular microenvironments at a

relatively larger scale than the sub-cellular level. This includes interaction of cells with

surrounding biomaterials14. Proper understanding of these interactions is essential

especially in fields such as tissue engineering and regenerative medicine. Manipulating

the cell environment using artificially engineered matrices is a primary focus in this field

of science15,168. Biomaterial environments provide a variety of physical, mechanical,

chemical as well as structural cues that can be altered to suit a specific application.

Page 77: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

77

Cardiovascular biomaterials in general encompasses a wide variety of material

applications ranging from vascular grafts to hemodialysis tubes. These biomaterials can

be designed to alleviate complications arising due to different cardiovascular diseases,

such as artherosclerosis, myocardial infarction, peripheral artery disease etc. The

current chapter and Chapter 5 will discuss two classes of cardiovascular biomaterial

applications, namely, small diameter vascular grafts and vascular stents, respectively.

This work is focused on studying the interaction of these biomaterials with in vitro

models of the vascular environment in order to understand and develop strategies to

repair damaged vasculature and enable regeneration.

With this goal in mind, the following aim was established and studied. Within the

broad scope of this aim, specific sub-goals were also listed to study the two classes of

biomaterial applications as described above.

Specific Aim 2

Understand and develop biomaterial based strategies towards vascular tissue

engineering and regeneration by studying the interactions of biomaterials with vascular

microenvironments.

Sub-aim 1

Develop a novel biomimetic, bilayered, biomaterial construct and evaluate its

applicability as a small diameter vascular graft.

Sub-aim 2

Evaluate the biocompatibility and hemocompatibility of an Mg-Ca-Sr alloy to

assess it suitability for use in designing vascular stents.

As mentioned above, this chapter will deal with the design and development of a

biomimetic vascular scaffold for small diameter blood vessel engineering. The field of

Page 78: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

78

vascular tissue engineering aims to create replacement vascular tissue for diseased or

damaged segments of the vasculature. The end goal of this research is to restore

adequate blood flow to regions of the body where blood flow is lacking because of

damage or diseased state of the existing vasculature, or in some cases, the complete

destruction of the vascular network6,8. Current efforts in the field of vascular tissue

engineering are focused on creating functional replacements for small diameter blood

vessels (<6mm). Furthermore, the golden standard for vascular grafts is the use of an

autologous vessel that would be biologically acceptable for the patient. But the

availability of autologous vessels is limited due to pre-existing conditions or failure to

identify or extract a vessel of appropriate size with minimal damage to the existing

system. These clinical limitations have steered research in this field towards developing

synthetic tissue engineered vascular grafts6,8,117. Current grafts are not completely

successful and often fail due to complications after implantation, such as mechanical

failure, thrombosis or increased inflammation. An ideal vascular graft should be

nonthrombogenic or thromboresistant, non-toxic, cause minimum inflammatory

response, ease of processing and handling, and mechanically compatible with existing

vasculature75. Regardless of whether the material used to create the vascular graft is

synthetic or natural, it is also essential that the scaffold can support vascular cell

adhesion and proliferation to enable reintegration into the vascular network. Although

there have been decades of advancement in vascular cell biology, there remains the

challenge of identifying a suitable material and an ideal graft design. Recent

approaches aim to achieve acceptable patency rates, through the development of grafts

Page 79: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

79

that best mimic or promote the extracellular environment and mechanical properties of

native blood vessels4,169.

The identification of a single material or a combination of materials for fabrication

of grafts has been a pivotal research focus in the field. A popular and widely practiced

approach is the use of proteins derived from plant and animal sources as the base

material for scaffold18,79,89,170,171. However, this method is limited by the low mechanical

strength of the scaffolds, high cost of proteins as well as difficulty in processing them.

Historically, many synthetic materials have been identified as suitable materials in

vascular graft design due to the ease and flexibility of tailoring their mechanical and

structural properties. Due to the poor patency rates of most synthetic material grafts,

recent efforts focusing on the use of biodegradable polymers as scaffolds to improve

the clinical viability have been made. In this design, the scaffold will degrade and be

replaced with extracellular matrix proteins that are secreted by the cells over time.

Some common materials that have been investigated include polyglycolic acid, poly-L-

lactic acid, and co-polymers of the two70,77,109. However, synthetic materials have

increased risk of thrombogenicity, toxicity, mechanical failure and inflammatory

response. Novel strategies including surface functionalization to impart greater anti-

thrombogenic properties of the material, via coatings, chemical and protein

modifications, and cellular seeding on these otherwise inert materials, have also been

attempted172–175. But the long-term toxicity as well as anti-thrombogenicity of purely

synthetic grafts are still not completely verified. These challenges have directed

research towards novel hybrid material systems that combine the positive

characteristics of natural as well as synthetic materials. This study will investigate the

Page 80: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

80

use of a combination of a biodegradable, elastomeric polymer; poly (1,8 octanediol-co-

citrate) (POC) with natural extracellular matrix proteins; collagen and elastin. POC

belongs to a family of elastomers composed of citric acid and an aliphatic diol. POC has

been identified as a hydrophilic and biodegradable polymeric constituent that can

substitute for elastin in synthetic fibres.80–83 It is also anti-thrombogenic and supports

differentiated endothelial progenitor cell adhesion growth.84,85

This polymer-protein mixture was fabricated into a fibrous construct using

electrospinning. Electrospinning offers robust material selection, low cost, simplicity,

high surface-to-volume ratios, and controlled porosity for vascular scaffold fabrication.

With these advantages, electrospinning is a proven framework allowing for the improved

endothelialization of vascular grafts89,96,170. While some encouraging data using

synthetic electrospun polymers have been obtained, these approaches are still limited

and some of the challenges pertaining to their lack of bioactive functionality are still

present97. Electrospinning of POC with natural co polymers and their suitability as

scaffolds for vascular grafts has been successfully demonstrated before81. This study is

focused on electrospinning POC with collagen to closely mimic the architecture of the

native vessel wall.

Apart from selection of appropriate biomaterials, microstructure as well

dimensions of the scaffold are an important criterion for success14,27,176. Current efforts

have focused on mimicking the native structure of the arteries to improve cellular

compatibility of the constructs by providing essential structural cues. The design of the

Page 81: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

81

construct in this study is of a scaffold structure consisting of two layers as shown in the

illustration below (Figure 4-1).

Figure 4-1. Schematic representation of the bilayered vascular graft.

Native artery as shown in Figure 2-3, consists of a solid inner layer known as the

lumen and a fibrous exterior layer also known as the medial layer. The inner luminal

layer is composed primarily of a monolayer of endothelial cells that are in direct contact

with circulating blood. The medial layer, consists of multiple layer of smooth muscle

cells. This layer is also rich in collagen and elastin among other ECM proteins and

provides a great amount of mechanical support to the vessel91,177.

The goal of this study was to mimic this structure of as closely as possible.

Therefore, the design as described above was chosen to support a monolayer of

endothelium and an interconnected multi-layer of SMCs. This report describes the

development of a bilayered, biomimetic scaffold. This strategy is currently being

developed to recruit stem cells to improve regeneration and reintegration into the native

vasculature.

Materials and Methods

Materials

POC pre- polymer was synthesized using a protocol reported by Yang et al. 178.

Briefly, equimolar ratios of 1, 8 octanediol (Sigma-Aldrich, St. Louis, MO) and citric acid

(Sigma-Aldrich, St. Louis, MO) were blended together and melted at 165˚C. Once the

Page 82: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

82

monomers were completely molten, the poly condensation reaction was carried out at

140˚C. The pre-polymer solution was then purified reconstituted at concentration of 50

wt.% in ethanol and 10 wt.% in 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) (Sigma-

Aldrich, St. Louis, MO) for lumen solvent casting and electrospinning respectively.

Lyophilized collagen type I from calf skin (Elastin Products Company, Inc., Owensville,

MO) and soluble elastin (Elastin Products Company, Inc., Owensville, MO) were used to

make the protein-polymer mixture for electrospinning. Polyacrylic acid (PAA) powder

was obtained from Sigma Aldrich and was reconstituted to 10 wt.% in ethanol.

Solid Lumen Fabrication

To fabricate a solid lumen consisting of POC, First, a 5 wt. % Polyethylene oxide

(Peo) solution in water was electrospun on an aluminum rod. The polymer solution was

fed through a 25G needle at 2 mL/ hr with a syringe pump (Chemyx Fusion 100,

Stafford, TX). A voltage potential of 25 kV was applied and the rod was placed 15 cm

away from the syringe tip. The rod was rotated at a speed of 500 RPM to ensure

uniform distribution of fibers The Peo spun rod was dipped in a 50 wt. % solution of

POC pre-polymer in ethanol and the solvent was evaporated overnight in air at room

temperature. To prevent irregular flowing of the pre-polymer from the rod while

subsequent fabrication steps are carried out, the POC coated rod was then partially

crosslinked at 60˚C and 80˚C for one day each.

Fabrication of Electrospun Medial Layer

10 wt. % solutions of Collagen, POC and Elastin in HFIP were prepared for

electrospinning. The collagen: POC: elastin solution was made with a 60:20:20 weight

ratio of each of the components respectively. The mixture was vortexed for 15 min

under low speed to thoroughly mix the constituents. The collagen: POC: elastin solution

Page 83: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

83

was fed through a 25G needle at 1.25 mL/ h with a syringe pump and a voltage

potential of 22-25 kV was used; the working distance was set at 15 cm.

Glutaraldehyde Crosslinking

The electrospun scaffolds were then crosslinked using glutaraldehyde vapor. The

scaffolds were crosslinked immediately after electrospinning to stabilize the proteins in

the scaffold and to improve mechanical strength of scaffold. The rods were suspended

in a set up as shown in schematic below (Figure 4-2 A). The cylinder contained 30 ml of

glutaraldehyde (25% in water). For flat sheets, a similar set up was shown where the

scaffold is placed in an enclosed setting (Figure 4-2 B). The scaffolds were crosslinked

for 2, 4, 6 and 8 h. After crosslinking the scaffolds were soaked in water to wash away

any residual glutaraldehyde and to enable demolding of the bilayered construct

Figure 4-2. Glutaraldehyde vapor crosslinking. (A) Electrospun rod crosslinking set up (B) Electrospun mat crosslinking set up.

A

B

Page 84: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

84

FTIR Characterization

The crosslinking of scaffolds was confirmed by FTIR. Spectro chemical analysis

of the electrospun scaffolds crosslinked at various times was performed using

attenuated total reflectance Fourier transformation infrared spectroscopy (ATR-FTIR).

ATR-FTIR spectra was obtained for each of the samples using the Nicolet 6700 FTIR

Spectrometer (Thermo Scientific) and a diamond tip window. The spectra were read

over a range of 600-4000 cm-1 for each of the spectra and a total of 32 scans were used

with a resolution of 4 cm-1.

SEM Characterization

The scaffolds were washed and frozen at -80˚C for 18 h before being lyophilized.

The dried scaffolds were then mounted for SEM analysis and the nanofibrous structure

of the scaffolds were observed using a table top SEM (PhenomWorld ProX, Eindhoven,

Netherlands). Fiber size distribution was quantified from SEM images using ImageJ

(National Institutes of Health Bethesda, MD).10 images taken at 10000 X magnification

were used and 15 measurements were taken from each of the images. The average of

the fiber size measurements was reported and a distribution was plotted.

Contact Angle Measurement

The contact angle of the solid POC lumen as well and the collagen: POC: elastin

layer were characterized using a custom goniometer (courtesy: Brennan lab, UF). Solid

films of POC and the 60:20:20 collagen: POC: elastin were fabricated by solvent casting

the polymer solutions on glass slides and solvent was allowed to evaporate overnight.

The films were crosslinked using the conditions reported earlier. The glass slides were

placed on the custom goniometer and a droplet of 5 µL of deionized water was placed

Page 85: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

85

on the film. This was repeated three times for each of the films. The contact angle

observed was averaged and reported and correlated with hydrophilic behavior.

Uniaxial Tensile Testing

Mechanical properties of the electrospun protein polymer construct was

determined using a tabletop uniaxial testing machine (INSTRON 3345) equipped with a

10-N load cell. A cross-head speed of 10 mm/min at room temperature was applied to

hydrated samples after being punched into dog bone shaped specimens. A sample size

of at least 5 was used for the testing procedure.

Biocompatibility Characterization

Cell culture

Human umbilical vein endothelial cells (HUVECs) (Lonza, Walkersville, MD) and

human aortic smooth muscle cells (hASMCs) (Cell Applications Inc., San Diego, CA)

were used as representative mature vascular cell populations to evaluate the response

of the different layers on the construct with relevant vascular cells. Both cell types were

cultured in a cell culture incubator maintained at 37˚C, 5% CO2. HUVECs were cultured

in endothelial growth media-2 (EGM-2) (Lonza) and were used between passages 3-5

and while the hASMCs were cultured in smooth muscle cell growth media (SmGM) (Cell

Applications Inc) and used between passages 8-12.

Scaffold preparation for cell studies

For biocompatibility evaluation, the scaffolds were punched into discs of diameter

(1/16”) following fabrication. The scaffolds were then gas sterilized with ethylene oxide

gas prior to use, in a gas sterilizer (Anaprolene AN741, Vernon Hills, IL). Unreacted

monomers and residual glutaraldehyde was leached from the scaffold using Dulbecco’s

Modified Eagle Medium (DMEM) (Corning Cell Grow, Mediatech Inc., Manassas, VA)

Page 86: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

86

for one week with constant replacement of medium. The scaffolds were then washed

with 1x phosphate buffered saline (PBS) and then incubated with cell culture medium

overnight before cell seeding.

Cellular attachment

To assess cellular attachment and spreading, appropriate cells were seeded on

the solid lumen layer and the electrospun medial layer separately. HUVECs were

seeded at a density of 5000 cells/mm2 on the POC discs and hASMCs were seeded at a

density of 5000 cells/mm2 on the collagen: POC: Elastin discs respectively. The cells

were seeded by placing a 50 µl cells suspension directly on the scaffold surface.

Special care was taken to avoid any spillage. The seeded scaffolds were then carefully

placed in a cell culture incubator and allowed to attach for 45 minutes. Following this

incubation, scaffolds containing wells were filled with about 300 µl of media. Cells were

then cultured on the scaffolds for two days. After two days of culture, scaffolds were

rinsed twice with PBS to remove any unattached cells, fixed with 2% glutaraldehyde.

The fixed scaffolds were the serially dehydrated with graded series of ethanol (30%,

50%, 70%, 90%, 95% and 100% ethanol in water) by incubating the scaffold in each

solution for 10 min. Scaffolds were then lyophilized, mounted, and coated with Au-Pd

via sputter coating for SEM analysis.

Lactate dehydrogenase cytotoxicity assay

Preliminary cytotoxic response of the scaffolds was evaluated by quantifying the

concentration of lactate dehydrogenase (LDH) released in cell culture supernatant

media. LDH cytotoxicity assay kit (Thermo Scientific, Waltman, MA) was used to

colorimetrically detect the concentration of LDH in the media. Scaffolds were placed in

24 well ultra-low attachment plates (Fisher Scientific, Waltman, MA). Briefly, 5000

Page 87: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

87

HUVECs were seeded onto the POC layer and 5000 hASMCs were seeded on the

fibrous scaffold layer cultured in 96 well plate with 100 µl of media in each well. The

optimal cell density for LDH release was previously assessed using a cell density vs

LDH release calibration curve. After two days of culture, the media was transferred into

a 96 well plate and assayed for LDH concentration. A subset of wells was treated with

lysis buffer provided to serve as a control for maximum LDH concentration. The relative

percentage cytotoxicity was reported per scaffold.

Cell proliferation assessment

The assessment of proliferation of cells on the medial layer consisting of

collagen: POC: Elastin, was studied using a 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl

tetrazolium bromide (MTT) assay (Trevigen, Gaithersburg, MD). The assay is designed

to spectrocolorimetrically indicate the metabolic activity of the cells through the

assessment of cellular ability to reduce the yellow tetrazolium salt to purple formazan

crystals by mitochondrial enzymes. Healthy proliferating cells can produce a greater

amount of crystals when compared to quiescent cells or cells undergoing apoptosis. For

proliferation assessment, cells were seeded as described in the LDH assay section

above. The initial cell seeding density was determined to be 2500 cells/mm2. Each well

was filled with 100 µl of cell culture medium. At time points of 1, 3, 5, and 7 days, 10 µl

of MTT reagent was added to media, allowed to react for 4 h, following which 100 µl of

detergent was added to solubilize the crystals produced overnight. The supernatant was

collected and the absorbance was measured using a plate reader. The absorbance

values were correlated with cell number using a previously determined standard curve

that correlated absorbance to cell number. The cell number versus culture time was

plotted.

Page 88: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

88

Optimizing Bilayer Fabrication with Electrospun POC-PAA

Once the biocompatibility and cellular compatibility of the electrospun scaffold

was confirmed. As the next step was focused on the fabrication of the bilayered

construct. The fabrication of the bilayer was optimized using an established

electrospinning system consisting of 10 wt. % POC: PAA (50:50) solution in ethanol.

This system has already been characterized and reported for wound healing

applications by the lab previously. The POC: PAA solution was electrospun on the

partially crosslinked lumen on the aluminum rod. The polymer solution was fed through

a 25 G needle at the rate of 2 mL/h. The collector plate distance was fixed at 20 cm and

the rod was rotated at 500 RPM. The fabricated construct was thermally crosslinked at

80˚C for 2 days and 100˚C for one day. The scaffolds were demolded from the rods by

soaking in water. The integration between layers as well as the fabrication of two

distinct phases was confirmed visually using SEM analysis of cross-sections of the

scaffold.

Fabrication of Bilayered Construct

To fabricate a bilayered construct, a layer of collagen: POC: elastin was spun on

a rotating mandrel consisting of the partially crosslinked lumen as described above. The

final biphasic scaffold was crosslinked in glutaraldehyde vapor for 6 h as determined

earlier. Once crosslinking was completed the scaffold was placed in deionized water for

two days then removed from the mandrel. The construct was also characterized using

SEM analysis as described above.

Page 89: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

89

Mechanical Properties

Compliance

Dynamic compliance of the scaffolds was measured using a custom bioreactor

apparatus (42 Bio, Gainesville, FL). Scaffolds were mounted on the metal adapters of

the custom bioreactor apparatus using UV glue, Loctite 3942 (Henkel Adhesives, Rocky

Hill, CT). Compliance was measured per ISO 7198 section 8.10 defined as the change

in volume divided by the change in pressure at the physiological diastole and systole

(approximately 120/ 80 mmHg). Using an LED micrometer (Keyence LS7000, Itasca, IL)

the change in outer diameter was measured. The following equation provided in the

ISO, Equation 4-1 was used to calculate the % compliance of the scaffolds.

% 𝑐𝑜𝑚𝑝𝑙𝑖𝑎𝑛𝑐𝑒 =

𝑅𝑝2−𝑅𝑝1

𝑅𝑝1

𝑝2−𝑝1× 104 (4-1)

Where Rp2 represents the radius at p2, the diastolic pressure and Rp1 represents the

radius at p1, the systolic pressure.

Circumferential hoop stress

Circumferential hoop stress was measured per ISO 7198 section 8.3. The

biphasic scaffolds were cut into ringlets, hydrated for 1 h, and loaded into stainless steel

hooks in a uniaxial mechanical testing apparatus (Instron 5942, Norwood, MA). Ringlet

width, wall thickness, and outer diameter were measured using a caliper for each

sample. A minimum of three measurements was taken per sample. The cross-head

speed was set at 50 mm/min, applying tensile force until failure of the scaffold was

observed. Load and displacement data were collected to determine the elastic modulus

of the scaffolds, using Equation 4-2 as shown below.

Page 90: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

90

𝐸 =𝜎

𝜀 (4-2)

The burst pressure of the scaffolds was extrapolated from the circumferential

hoop stress using the Laplace law which states burst pressure increases linearly with

decreasing diameter if the wall thickness is assumed to be constant. Equation 4-3,

where Pburst was the burst pressure, σy was the yield stress, t was wall thickness, and r

was the radius of the scaffold was used to calculate extrapolated burst pressure.

𝑃𝑏𝑢𝑟𝑠𝑡 =𝜎𝑦×𝑡

𝑟 (4-3)

Statistical Analysis

All numerical data are reported as average ± standard deviation. Statistical

significance was determined using one-way analysis of variance test with Fisher’s LSD

as the post hoc analysis. P values < 0.05 were reported as significant while P> 0.05

were reported as statistically similar.

Results and Discussion

Despite immense efforts, the search for a fully functional scaffold system that

enhances vascular regeneration is still a major focus of this field. One of the major

challenges has been the identification of a suitable material to develop small diameter

vascular graft replacements. Historically, many synthetic as well as natural materials

have been utilized for the fabrication of vascular grafts. Additionally, various changes to

the processing conditions, surface properties and material geometry have also been

investigated towards the development of patent and compliant vascular grafts for small

diameter vessel applications. Even though natural materials, are most suited for these

Page 91: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

91

applications due to their bio functionality, the weak mechanical properties as well as the

high cost and difficulty in processing limits their application. Synthetic materials despite

having high mechanical properties have limited bioactivity. Therefore, the use of a

hybrid protein-polymer system has been evaluated in this study.

The overall goal of this study to fabricate a bilayered biomimetic scaffold for

potential application as a vascular graft. The rationale behind this approach is to

develop a scaffold system that mimics architectural and functional aspects of an existing

arterial environment to promote vascular cell attachment and growth. The design as

shown in Figure 4-1 consists of solid inner layer consisting of POC and a fibrous

electrospun exterior layer consisting of POC combined with ECM proteins (collagen and

elastin). POC belongs to an elastomeric family of synthetic polymers, termed poly (diol

citrates) (PDC) composed of citric acid and an aliphatic diol. The condensation reaction

involved in the synthesis of polydiol citrates is shown below in Figure 4-3.

Figure 4-3. Synthesis of poly diol citrates.

Amongst these elastomeric polymers poly (1, 8- octane diol) citrate (POC) has

been identified as a hydrophilic and biodegradable polymeric constituent that can

substitute for elastin in synthetic fibers. It is also anti-thrombogenic and supports

endothelial progenitor cell adhesion growth which is required for the blood contacting

lumen of the graft83,179. Therefore, this material was chosen to form the base lumen

layer to support growth of a neoendothelium. This layer was fabricated by solvent

Citric acid Poly diol

Page 92: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

92

casting a viscous solution of POC in ethanol and partially crosslinking the layer to

preserve the structure of the lumen during subsequent processing stages.

The idea behind fabrication of the bilayered scaffold was to fabricate by solvent

casting a solid POC lumen onto a mandrel and electrospinning a nanofibrous collagen:

POC: elastin layer on top of the lumen. Towards this goal, an electrospun composite

consisting of collagen, POC, elastin was first fabricated. Electrospinning offers robust

material selection, low cost, simplicity, high surface-to-volume ratios, and controlled

porosity for vascular scaffold fabrication. With these advantages, electrospinning is a

proven framework allowing for the improved reintegration of vascular grafts. While some

encouraging data using synthetic electrospun polymers have been obtained, these

approaches are still limited and some of the challenges pertaining to their lack of

bioactive functionality are still present97. Electrospinning of POC with natural co

polymers and their suitability as scaffolds for vascular grafts has been successfully

demonstrated before79,81,180–182. Natural ECM proteins, collagen and elastin have been

known to be present in the ratio of 80:20 in native arteries177,181,183. This ratio has been

adopted while designing collagen and elastin based vascular scaffolds68,184,185. In this

study, the idea is to substitute a part of the collagen for elastomeric POC to improve the

mechanical robustness and compliance of the scaffold. Therefore, collagen: POC:

elastin was mixed in a ratio of 60 : 20 :20 by weight and dissolved in a common solvent

HFIP to electrospin the mixture.

Page 93: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

93

The electrospun mixture produced uniform fibers of size 0.816 nm as shown in

Figure 4-4 below and the electrospun scaffolds were water soluble and had to undergo

chemical crosslinking with glutaraldehyde vapors to stabilize the proteins.

Figure 4-4. SEM characterization as spun collagen: POC: elastin fibers. (A) SEM image of as spun Collagen POC Elastin fibers, 5000X magnification (B) Fiber size distribution analysis.

Glutaraldehyde has been used historically as a strong crosslinking agent to

crosslink and stabilize proteins186. It reacts with the amino groups present in the

proteins to form Schiff base intermediates which then link further to form a large

crosslinked network. Glutaraldehyde crosslinking has been shown to be a cost effective,

fast and an efficient process which increases the mechanical strength and denaturing

temperature of the crosslinked proteins. However, some studies have shown that this

process can leave behind unreacted aldehydes, which when released causes local

toxicity to surrounding tissue187–189. Therefore, care needs to be taken to remove

unreacted aldehydes released by this process.

A B

Page 94: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

94

In this study, the scaffolds were crosslinked at various crosslinking times to

determine an optimal crosslinking time and their stability was preliminarily tested by

observing their behavior when immersed in water. Scaffolds crosslinked for shorter

times, less than 1 h seemed to readily dissolve in water. Therefore, in this study,

scaffolds were crosslinked for 2, 4, 6 and 8 h. The crosslinked scaffolds were all stable

in water and retained their structural characteristics of crosslinked fibers as shown in the

Figure 4-5 below.

Figure 4-5. SEM characterization of all crosslinking conditions using glutaraldehyde vapor. All images are at a magnification of 5000 X.

Further FTIR analysis confirmed the crosslinking of scaffolds as indicated by a

characteristic Amide II (NH) bending vibrations peak observed at around 1550 cm-1.

Additionally, Amide I (C=O stretching) and Amide A (N-H stretching) signatures were

also seen respectively at 1632- 1664 and 3320-3340 cm-1 for all scaffolds tested. As

Page 95: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

95

time of crosslinking increased, an increase in peak absorbance was observed in the

Amide bands indicating increase in crosslinking (Figure 4-6).

Figure 4-6. ATR-FTIR spectra of collagen: POC: elastin crosslinked fibers.

Table 4-1. Average fiber diameter of collagen: POC: elastin fibers with different cross-linking times.

Time of crosslinking Average fiber diameter (µm)

Uncrosslinked 0.816 2h 1.044 4h 1.436 6h 1.900 8h 2.414

Higher magnification SEM images of the nanofibrous layer revealed a uniform

bead-less nanofibrous network with microporous structures. Further SEM

characterization of the scaffolds showed that with higher crosslinking times the

openness and apparent porosity between fibers decreased. The average fiber diameter

of the scaffolds also increased with higher crosslinking time as shown in Table 4-1

above. The average fiber diameters ranged from 0.816 µm-2.414 µm. Previous work by

Rüder et al. 2013 demonstrated significant smooth muscle cell adhesion, spreading,

and proliferation on scaffolds with submicron sized fibers 190. As discussed in previous

Page 96: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

96

chapters, porosity and openness are important characteristics for adequate cell growth.

Therefore, to balance the openness of structure as well as to ensure acceptable

mechanical compatibility, a 6h crosslinking time was chosen for scaffolds prepared for

the rest of this study.

Another important physical characteristic of biomaterial scaffolds is their

hydrophilic nature. Many studies in the past have shown that a hydrophilic surface is

preferred for biomedical applications. The wettability of the surface plays an important

role in cell adhesion and spreading, thereby influencing the biocompatibility of the

biomaterial191–193. To assess the hydrophilicity of the surfaces used in this study, the

contact angles of the water – biomaterial interface on POC and collagen: POC: elastin

films were tested. The experiment showed that contact angle at the sample water

interface on POC and collagen: POC: elastin films, were 60.78 ± 5.94° and 55.62 ±

8.12° respectively. The image of the water droplet and the contact angle measure are

shown below in Figure 4-7 and Table 4-2 below. The fact that both contact angles were

below 90° implied that both layers of the biphasic scaffold exhibited hydrophilic nature

and therefore are suitable for cell adhesion.

Figure 4-7. Contact angle measurements. A representative digital image of a water droplet on (A) collagen: POC: elastin film and (B) POC film.

Table 4-2. Contact angles at the water – biomaterial interface

Material Composition Contact Angle

Collagen: POC: Elastin 55.62˚ ± 8.12˚

POC 60.78˚ ± 5.94˚

A B

Page 97: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

97

The medial layer of native arteries are enriched with collagen and have been

known to carry the majority of the mechanical load in an artery90,177,194. It was essential

that the collagen: POC: elastin layer was able to provide the required mechanical

support to the construct. The preliminary mechanical properties (as shown in table 4-3

below) of the electrospun scaffolds were measured by quantifying its uniaxial tensile

properties. The hydrated tensile testing of the scaffolds showed a young’s modulus

value of 1.89 +/- 0.5 MPa and UTS value of 0.76 +/- 0.04 MPa. These results align with

values reported for medial layer of human carotid artery in literature195–198. Further the

percentage elongation of the scaffold was measured to be 60.9 +/- 16.34 %. Although

this value was not comparable to arterial elongation in vivo, the addition of an

elastomeric POC layer is expected to improve the elasticity of the scaffold construct.

Table 4-3. Uniaxial tensile test. Assessment of tensile properties of the glutaraldehyde vapor crosslinked electrospun mat

Young’s modulus (MPa)

Ultimate Tensile Strength (MPa) % Elasticity

1.89 +/- 0.5 0.76 +/- 0.04 60.9 +/- 16.34

An essential requirement of a cardiovascular biomaterial is biocompatibility and

capability to house appropriate vascular cells and to promote their growth. It has been

shown the incorporation of natural ECM components like collagen and elastin in tissue

scaffolds provide a biomimetic substrate for scaffolds and promote cell functions

relevant for tissue regeneration78,81,89,184. Preliminary biocompatibility of the scaffold

construct was assessed by evaluating cell adhesion and proliferation on both layers of

the bilayered scaffold. Additionally, the cytotoxic response invoked by the scaffold

layers was also quantified using an LDH assay as described in the methods section.

Page 98: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

98

Previous reports have raised concerns about potential cytotoxic risk posed by

residual aldehydes from the glutaraldehyde crosslinking of proteins. Therefore, in this

study, the electrospun and glutaraldehyde vapor crosslinked scaffolds were leached by

soaking in DMEM for at least a week before use in biocompatibility experiments.

HUVECs and hASMCs were used to perform biocompatibility assessments because a

monolayer of endothelial cells makes up a significant portion of the intimal layer in

native vasculature and smooth muscle cells reside in the nanofibrous medial layer.

Corroborating the hypothesis from the contact angle measurements and previous work

that has reported adhesion of endothelial cells on POC films 178,179,199,200.

Figure 4-8. SEM images of cell attachment. hASMCs seeded onto collagen: POC: elastin fibers.

SEM imaging (Figure 4-8) revealed that the hASMCs adhered to the collagen:

POC: elastin and show preliminary evidence of migration under the electrospun fibers

into the bulk of the scaffold. But due to the presence of proteins in the matrix, fixation of

cells with glutaraldehyde introduced some artifacts in the SEM image as shown above.

Therefore, the morphology of the fibrous scaffold was not clearly visible. Current efforts

to image the cell seeded scaffolds using alternate techniques such as environmental

Page 99: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

99

SEM or confocal microscopy is underway. Nevertheless, the images indicated

preliminary evidence of cell attachment and spreading indicating healthy cell adhesion

on scaffold layers. These results were in accordance to enhanced smooth muscle cell

adhesion on collagen, elastin, and POC individually and in combination with other

materials as shown in literature.90,178,200,201

Figure 4-9. Relative cytotoxicity measurements of collagen: POC: elastin with hASMCs and POC lumen with HUVECs. Data are reported as mean ± standard deviation (n=3). N.S. indicates no statistically significant difference (p > 0.05) and * indicates a statistically significant increase (p < 0.05).

Colorimetric detection of the intracellular enzyme LDH was used to assess

cytotoxicity of the scaffold layers on the HUVECs and hASMCs. Relative cytotoxicity

measurements revealed a 5.83 ± 3.68 % cytotoxicity for the HUVECs on the POC

lumen and 5.49 ± 0.91 % on the tissue culture plastic. For the hASMCs the cytotoxicity

of the hASMCs on the collagen: POC: elastin electrospun layer was 14.24 ± 7.96 %

compared to 21.18 ± 11.12 % on tissue culture plastic (Figure 4-9). Compared to the

tissue culture plastic control there was no statistical significance (p > 0.05) in the

cytotoxicity of the two layers of the bilayered scaffold.

Page 100: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

100

After confirming the preliminary biocompatibility of the scaffold layers, the

proliferation of cells on the electrospun medial layer was assessed via metabolic activity

which was correlated to cell number. At time points of 1, 3, 5, and 7 days MTT reagent

was added and then the purple formazan crystal produced were solubilized. The

absorbance of the media was read for the hASMCs on the collagen: POC: elastin

(Figure 4-10).

Figure 4-10. Proliferation kinetics. Kinetic curve representing the proliferation of hASMCs on the collagen: POC: elastin. Data are reported as mean ± standard deviation (n=3). N.S. indicates no statistically significant difference (p > 0.05) and * indicates a statistically significant increase (p<0.05).

After the initial seeding at t = 1 day the cell number was calculated as 7133.33 ±

321.46 cells. At t = 3 days and t = 5 days the cell number increased to 7566.67 ± 57.73

and 8300 ± 264.58 cells respectively. From t =3 days to t = 5 days there was a

statistically significant increase in cell number (p < 0.05). At t = 7 days, the extrapolated

cell number was 9933.33 ± 513.16 and between t = 5 days and t = 7 days there was a

statistically significant increase (p < 0.05).

Page 101: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

101

The biocompatibility results indicate the scaffold is able to support relevant cell types

growth and proliferation and preliminarily promote relevant cell functions to support

tissue regeneration. To fully assess the regenerative capacity of the scaffold further

studies would need to be done with longer culture times and in a dynamic environment

or in vivo.

Figure 4-11. SEM characterization of POC; POC-PAA bilayered construct. (A) the biphasic scaffold that contains solid lumen and nanofibrous medial layer, 500X (B) interface between solid lumen and electrospun fibers, 2000X, and (C) the fibrous electrospun mat, 2000X.

Encouraged by these preliminary results, the next step was the optimization of

the bilayered construct fabrication. The methodology of this process was optimized

using a solid lumen layer consisting of POC surrounded by an electrospun POC: PAA

layer. The POC layer was partially thermally crosslinked for 2 days at 60 ̊C and 80 ̊C

respectively. A layer of POC PAA was then electrospun onto the POC layer using

predetermined electrospinning parameters. The scaffold construct was further

crosslinked thermally as described. The fabricated scaffold was then visually inspected

using SEM imaging (Figure 4-11) which showed complete integration between the two

layers and the clear distinction between the porous morphology of the medial

electrospun layer versus the solid non-porous POC luminal layer.

Page 102: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

102

The fabrication technique optimized above was applied to construct the bilayered

scaffold incorporating a medial layer of electrospun collagen: POC: Elastin. The

fabricated construct was also crosslinked for 6h in gluataraldehyde vapor and

crossections of the scaffold was SEM imaged. Images (Figure 4-12) also revealed

integration between the electrospun and solid polymer layer of the biphasic scaffold.

Figure 4-12. SEM characterization of POC; collagen: POC: Elastin bilayered construct. Representative SEM image of (a) the biphasic scaffold that contains solid lumen and nanofibrous medial layer, 500X (b) interface between solid lumen and electrospun fibers, 2000X, and (c) the fibrous electrospun mat, 2000X.

The mismatch in compliance between a vascular graft and its host artery is one

of the major reasons for failure after implantation. Compliance mismatch has been

known to cause (1) anastomotic rupture, (2) flow induced shear stress and, (3) reduced

distal perfusion. All of these, factors eventually lead to build up of uneven stresses and

affect long term patency of the grafts202–204. Therefore a compliant scaffold that closely

matches the mechanical properties of native vasculature is imperative to successfully

design a tissue engineered vascular construct 8,9,87. The assessment of the

circumferential mechanical properties of the bilayered construct revealed highly

promising results. Salacinski et al., showed based on literature values there is a linear

relationship between the patency of the scaffolds and how closely the compliance

Page 103: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

103

matches native arteries. In this study, the saphenous vein, an autologous replacement

with the most similar compliance(4.4 ± 0.8 %), compared to native arteries(5.9 ± 0.5%),

showed the greatest patency rate (75%).However, ePTFE, a synthetic graft material

with a significantly higher compliance, (1.6 ± 0.2 %) exhibited the lowest patency rate,

40% 205.

Table 4-4. Circumferential mechanical testing of bilayered scaffolds compared to native tissue and ePTFE. 202,203,206,207.

Bilayered Scaffold Native Human Artery

Human Saphenous Vein ePTFE

Compliance (%) 6.9 ± 0.14 5.9 ± 0.5 4.4 ± 0.8 1.6 ± 0.2

Burst Pressure (mmHg) 401.68 ± 125.31 3,196 ± 126 1,599 ± 877 -------

Young’s Modulus (MPa) 0.330 ± 0.08 9-12 2.25-4.2 2.2

Circumferential Tensile Strength (MPa)

0.201 ± 0.06 ------ 3.7 ± 2.0 -------

The mechanical compliance (as shown in Table 4-4) of the scaffold was 6.94 ±

0.14% which is very close to reported values for native vasculature and therefore has

the potential to maintain a good patency rate similar to that of the saphenous vein,

probably even better than the current clinical synthetic standard, ePTFE202,204,205.

Further, the Young’s modulus of the bilayered scaffold was calculated to be

0.330 ± 0.08 MPa, and the burst pressure extrapolated using the Laplace relationship

was 401.68 ± 125.31 mmHg. The theoretical burst pressure was lower than native

arteries, (3,196 ± 126 mmHg), and saphenous veins (1,599 ± 877 mmHg)207. Although,

the bursting pressure was more than physiological levels of pressure, it is hypothesized

Page 104: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

104

that as the cells infiltrate and deposition ECM, they actively begin remodeling the

scaffold underneath. This may potentially lead to improved mechanical properties such

as Young’s modulus and burst pressure that align more with that of native tissue.

Niklason et al. showed from 5 to 7 weeks PGA scaffolds cultured dynamically with

vascular cells showed a remarkable increase in burst pressure from approximately 600

mmHg to 2200 mmHg117,206.

The main goal of designing tissue engineered vascular grafts is to find a

functional replacement for native vessels. These vessels consist of a dynamic,

hierarchical and complex structure and composition. In order to create a successful

replacement for native vessels, the design strategy to try to mimic not only the

composition but also the microarchitecture of the native vessel. The idea behind this

strategy is to best imitate the microarchitectural, chemical as well mechanical cues

provided by the ECM to a native blood vessel. With this rationale in mind, this study

involved design, fabrication and characterization of a novel bilayered biomimetic

construct consisting of a solid lumen and a fibrous medial layer. The medial layer was

enriched with native ECM proteins to improve bioactivity of the scaffold system. Overall

the results of this study indicate show that the bilayered scaffold was preserved the

architectural characteristics as shown by SEM imaging. It was also biocompatible and

was able to promote adhesion and proliferation of relevant vascular cell types.

More importantly the mechanical properties of the construct were compatible to

existing autologous replacements which are the golden standard in the field currently.

This scaffold is currently being developed to recruit circulating progenitor cells from the

blood and use them to endothelialize the graft surface. It is also believed that this

Page 105: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

105

strategy will promote integration of the scaffold into existing vasculature. Although many

previous reports have demonstrated the use of electrospun protein-polymer blends for

vascular grafts, mechanical compliance comparable to native tissue as well improved

biocompatibility has not been reported so far. This novel scaffold system is a promising

strategy for developing readily available vascular graft implants.

Page 106: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

106

CHAPTER 5 MESOSCALE STRUCTURAL CUES- PART 2- HEMOCOMPATIBILITY EVALUATION

OF A NOVEL MG-CA-SR ALLOY FOR VASCULAR STENT APPLICATIONS

Introduction

Another important class of cardiovascular biomaterials include metals which are

majorly used in designing heart valves, endovascular stents, and stent-graft

combinations. Metallic biomaterials are largely used in biomedical applications due to

their inertness and high mechanical strength67. However, some of the commonly used

bio-metals possess limited biofunctionalities, such as blood compatibility, bone

conductivity and bioactivity. To be used in cardiovascular applications, bloody

compatibility as well as compatibility with surrounding cellular environment (based on

the application), is a must. Failure of which leads to complications such as thrombosis

and inflammation which not only prevents incorporation of the implant into the existing

vasculature but also causes significant damage to it208,209. Therefore, the goal of

designing implants for vascular engineering is to design implants with cues that enable

repair of existing vasculature and prevent any susceptibility to damage due to the

implant placement.

As mentioned in the earlier chapters, the interaction of a biomaterial with its

surrounding environment is determined by its composition as well as structural features.

These factors govern the immediate as well as long-term responses after implantation.

In case of bio-inert metallic implants, a very thin layer of metal oxide formed, provides

the ultimate interface between stent and host tissue. The type of metal oxide formed

itself varies based on the composition of the material and the processing conditions

used. It has also been known that the thrombogenic response as well as tissue

Page 107: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

107

incorporation is largely determined by physical characteristics of the surface, such as

roughness, electrical change, free surface energy, and wettability101,103,107.

The initial interaction of metallic implants with the surroundings and dynamic

series of events that immediately follow triggers responses that facilitate the growth of

endothelial cells over the surface of the prosthesis107. Similarly, biomechanical behavior

also seems to affect the host’s response as well. Therefore, it is important to study and

understand the physical and chemical characteristics of metallic biomaterials before

they can be used for blood-contacting applications. This will be essential to design

structural, mechanical and chemical cues presented by the implant to the surrounding

system to elicit a favorable response.

Atherosclerosis is a life threatening cardiovascular disease that causes the

formation of plaques in arteries and is characterized by inflammatory infiltrates, lipid

accumulation, cell death and fibrosis91,210,211. To treat this condition various procedures

have been employed to mechanically improve perfusion in the artery without resorting to

surgery98,209,212. The most common procedure is percutaneous transluminal coronary

angioplasty, by implantation of an intracoronary stent to expand the artery. Despite

major breakthroughs and advancements in the research on permanent vascular stents

for treatment of coronary arterial diseases, major challenges such as stent thrombosis,

size mismatch, mechanical blockages and late development of restenosis and

malposition still exist213. These challenges have motivated and steered the research in

this field towards biodegradable and bioabsorbable stent materials. Biodegradable

vascular stents are currently the focus of research and clinical advancement, due to 1)

their ability to support the growth and remodeling of native vascular tissue; 2) the

Page 108: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

108

possibility to deliver therapeutic agents through degradation; and 3) their temporary

nature which eliminates the risk of late stent thrombosis213–216. Historically many

polymeric biomaterials such as, Poly L-lactic acid (PLLA), polyglycolic acid (PGA),

polycaprolactone (PCL) and poly (D, L-lactide/glycolide) copolymer (PDLA) have been

investigated as candidate materials for biodegradable stent applications 70,210,214,215.

While polymeric stents are advantageous because of their increased ability for targeted

drug delivery, higher drug loading capacity, and potential ability to allow percutaneous

revascularization and surgical intervention when necessary, there are some significant

disadvantages to the use of polymeric stents. The major disadvantage being their low

mechanical strength which may lead to stent fracture and migration over time77,216,217.

Moreover, polymeric stents have been known to cause a significant degree of local

inflammatory response and the long term biocompatibility of the degradation products of

polymeric materials remain unclear70,108.

These limitations have caused increased interest in biodegradable metallic stents

in the recent years, including magnesium (Mg)-based materials210,218. Magnesium-

based materials have been investigated for a variety of biomedical applications,

including but not limited to orthopedic and cardiovascular implants111,219–223. This

approach is supported by the fact that Mg is a naturally occurring element in the human

body, which is the second most abundant cation after sodium and has been reported to

exhibit very low to no toxicity219. When considering the use of Mg in vascular stent

applications, early studies have shown the beneficial effect of Mg ions to treat coronary

artery disease based upon their inherent anti-thrombogenic properties114. Mg was

shown to inhibit platelet activation through modulating the release of platelet activating

Page 109: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

109

factors, tromboxane A2 and prostaglandin224–228. The challenge in using pure Mg in

vascular stent applications is its very high degradation rate which results in a significant

loss of mechanical integrity, which is of critical importance for vascular stent

applications. This challenge can be overcome by alloying Mg with elements such as

Aluminum, Zinc, Yttrium and Cerium to decrease the degradation rate. While successful

in decreasing the rate of degradation, these alloys are inappropriate for biomedical

applications due to increased toxicity219. Herein, an alloy composed of Mg, Calcium and

Strontium (Mg-Ca-Sr) as a candidate material for vascular stent applications was

investigated. The high strength and low degradation rate of this alloy composition was

reported by Berglund et al. 2012222. These studies also characterized and reported the

low cytotoxicity and bioactivity of the degradation products from this alloy. Although

good biocompatibility and low degradation rate are primary requirements for materials

for stent applications, the compatibility of the material with blood and blood components

is of utmost importance213. Hemocompatibility of a vascular stent material is essential to

prevent fatal complications such as thrombosis, restenosis and inflammation post

implantation. Therefore, this study focuses on evaluating the hemocompatibility of the

Mg-Ca-Sr alloy with whole porcine blood and blood components as well as human

vascular and blood cells. The interaction of the alloy with porcine blood was evaluated

via assessment of hemolysis, whole blood clotting, platelet adhesion and activation.

Additionally, the inflammatory response of the alloy was assessed by quantifying the

release of pro-inflammatory cytokines/chemokines such as Interleukin 1 beta (IL-1β),

Interleukin 6 (IL-6), Interleukin 8 ( IL-8) & Tumor Necrosis factor alpha (TNFα) by a

human monocytic cell line (THP-1 cells) when in contact with the alloy. Lastly, the

Page 110: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

110

cytotoxicity of the alloy degradation products on vascular wall specific endothelial cells

was evaluated. This work represents the first report providing a comprehensive

evaluation of the hemocompatibility of this Mg-Ca-Sr alloy, and is the foundation for

future work on the use of this alloy material for vascular stent applications.

Materials and Methods

Alloy Synthesis and Sample Preparation

Pure elements (98.5 wt. % Mg, 1.0 wt.% Ca, and 0.5 wt.% Sr) were melted in an

induction furnace and direct chill cast to cylindrical billets 60 mm in diameter and 120

mm in length. The billet was homogenized at 450°C for 24 h under inert atmosphere. It

was then machined to 50 mm diameter and extruded in a 1.5 MN press at 0.5 mm/s to a

diameter of 10 mm for an extrusion ratio of 1:25. Temperature of 275ºC was maintained

by induction heating. Samples for cytotoxicity and hemocompatibility studies were

fabricated by cutting discs ~1mm thick on a low speed cutting saw, then grinding at 320

then 1200 grit SiC paper on the faces and sides. Controls of medical grade 316

stainless steel were fabricated in a similar fashion, then passivated per ASTM

A967/A967M-13 in 30% nitric acid for 1 the at room temperature. Poly(L-lactide) (PLLA)

discs were also included as a control for degradable polymeric material. The discs were

made by solution casting 5 % PLLA solution in chloroform. The cast film was washed 5

times with ethanol and De-ionised water to remove any residual chloroform.

Cytotoxicity Evaluation

Human umbilical vein endothelial cells (HUVECs) (Lonza, Walkersville, MD) were

used as a positive control for mature endothelial cell functionality. Endothelial cell basal

media-2 (EBM-2) (Lonza) supplemented with 2% fetal bovine serum, human fibroblast

growth factor (hFGF), vascular endothelial growth factor (VEGF), human epidermal

Page 111: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

111

growth factor (hEGF), human recombinant insulin-like growth factor (R3-IGF-1),

ascorbic acid, gentamicin, amphotericin, and heparin is referred to as endothelial cell

growth media-2 (EGM-2). HUVECs were cultured in EGM-2 culture medium and utilized

within passages 3-5. To assess the cytotoxic risk posed by the alloy degradation

products to the surrounding cellular environment, sample extract medium was prepared

according to ISO 10993-5 where 1 mL of extraction medium was added per 3 cm2 of

sample surface area. Briefly, 9.5mm diameter discs of Mg-Ca-Sr, S.S, and PLLA were

sterilized with 70% ethanol followed by UV radiation for 1 hr. The sterilized samples

were then placed in phosphate buffered saline (PBS) for 24 h, followed by another 24 h

incubation at 37̊C and 5% CO2 in complete EGM-2 media. After which, the media

extract containing degradation products was collected. Following collection, the cell

culture supernatant was serially diluted to 10, 30 and 50% in complete medium and

assessed for LDH release according to a modified protocol reported by Berglund et

al.222 . The cytotoxic response of HUVEC to alloy degradation products was evaluated

by quantifying release of lactate dehydrogenase (LDH) in the culture medium. LDH

released into the media from damaged cells is a biomarker for cellular cytotoxicity and

cytolysis. Briefly, HUVECs were seeded at a density of 5000 cells/well of a 96 well plate

and allowed to attach for 18 h. The culture medium in the wells were then replaced with

alloy extract and incubated for 24 h in a cell culture incubator. Following manufacturer’s

protocol, a colorimetric substrate for LDH (Pierce Biotechnology, Rockford, IL) was

incubated with the extracts for 30 min at 37 ̊C. The reaction was stopped with the

addition of 1N hydrochloric acid. The optical density was measured at 490 nm with a

reference wavelength of 650 nm.

Page 112: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

112

Hemolysis

The percentage of red blood cell lysis (RBC) or hemolysis was quantified using a

modified protocol reported by Motlagh et al. 2006. Circulating peripheral blood was

collected from the exterior jugular vein of domestic pigs (Department of Animal

Sciences, University of Florida) in the presence of acid citrate dextrose anticoagulant.

All blood collection procedures were performed in accordance with the rules and

regulations of the Institutional Animal Care and Use Committee (IACUC) at the

University of Florida. To assess hemolysis, 9.5 mm diameter discs of the Mg-Ca-Sr

alloy, S.S, and PLLA were immersed in PBS overnight, then placed in 24 well plates.

The anticoagulated blood was then diluted 1:4 in 0.9% saline, then 1mL of diluted blood

was added to the wells containing test samples and incubated under gentle agitation on

a shaker for 2 h at 37 C̊. As a positive control for complete RBC lysis, whole blood was

diluted in distilled water an incubated in the absence of any of the test materials. Saline

diluted blood also incubated in the absence of the test materials serves as a negative

control for no lysis. The diluted blood from each well was centrifuged at 1000 X g for 10

min to pellet the intact RBCs. The supernatant, containing the released hemoglobin was

transferred to a 96-well plate in triplicates then the absorbance was measured at 545

nm. The percentage hemolysis was then calculated using the following formula shown

in below in Equation 4-1 (abs denotes absorbance).

%𝐻𝑒𝑚𝑜𝑙𝑦𝑠𝑖𝑠 =(𝐴𝑏𝑠 𝑜𝑓 𝑡𝑒𝑠𝑡 𝑚𝑎𝑡𝑒𝑟𝑖𝑎𝑙)−(𝐴𝑏𝑠 𝑜𝑓 𝑛𝑒𝑔𝑎𝑡𝑖𝑣𝑒 𝑐𝑜𝑛𝑡𝑟𝑜𝑙)

(𝐴𝑏𝑠 𝑜𝑓 𝑝𝑜𝑠𝑖𝑡𝑖𝑣𝑒 𝑐𝑜𝑛𝑡𝑟𝑜𝑙)−(𝐴𝑏𝑠 𝑜𝑓 𝑛𝑒𝑔𝑎𝑡𝑖𝑣𝑒 𝑐𝑜𝑛𝑡𝑟𝑜𝑙)𝑋100 (4-1)

Page 113: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

113

Evaluation of Platelet Adhesion and Activation

Platelets isolated from porcine whole blood was prepared using a modified protocol

as reported by Motlagh et al. (2006)229 to assess and quantify platelet adhesion and

activation in response to the Mg-Ca-Sr alloy in comparison with control materials, S.S,

and PLLA. Collagen coated plastic was included as a positive control for platelet

adhesion and activation 84. To assess platelet adhesion, platelet rich plasma (PRP) and

quantified platelets by measuring LDH released by platelets upon lysis was obtained.

Briefly, whole blood was centrifuged at 250 X g for 15 min and the platelet-rich

supernatant was removed and the number of platelets were counted using a

hemocytometer. Samples of the alloy, S.S and PLLA (9.5mm diameter) immersed in

PBS overnight, were incubated with 100 µl of PRP containing 1 X 106 platelets per

sample for 1h at 37 ̊C. The platelets adhered on the samples were first lysed by

incubation with 2% Triton-PBS buffer for 45 min at 37 ̊C. The number of adherent

platelets was determined by detecting the amount of LDH present after cell lysis

following manufacturer’s protocol. A calibration curve was generated from a series of

serial dilutions of known platelet concentrations and was used to determine the number

of adhered platelets.

The activation of platelets on exposure to alloy samples and control materials was

assessed by quantifying the release of platelet factor 4 (PF4) by activated platelets.

Briefly, Mg-Ca-Sr alloy, S.S and PLLA samples were incubated with PBS overnight. The

samples and controls were then incubated with 100 µl of PRP containing ~6 X 106

platelets, for 2h at 37̊C. The PRP was then collected and assayed to quantify the

amount of PF4 released via an ELISA (ABclonal Science, Inc., Woburn, MA) per

manufacturer protocol.

Page 114: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

114

Whole Blood Clotting Assessment

The clotting behavior of recalcified whole blood in the presence of the Mg-Ca-Sr

alloy, S.S and PLLA was assessed using a modified protocol previously reported

elsewhere. 84,229–231 Briefly, whole blood collected in the presence of anticoagulant was

added onto 9.5 mm sample discs in a multiwell plate (20 µl per sample/control).

Following addition of blood, 10 µl of 0.2 M CaCl2 was added to recalcify the blood to

activate the clotting cascade. The addition of CaCl2 was omitted for a subset of wells as

a control for no clotting. At 1, 30 and 60 min after the addition of CaCl2, 1 mL of distilled

water was gently added to each well and mixed to lyse free RBCs that were not part of

the clot formed. The liquid was then removed from each well and the absorbance of the

supernatant was measured. Higher absorbance indicated higher thromboresistance of

the material. The absorbance at 30 and 60 min from each sample/control was recorded

and was normalized to initial absorbance at 2 min and the percentage decrease in

absorbance was calculated and correlated with percentage increase in clotting. In this

study, glass served as a positive control representing the kinetics of uninhibited whole

blood clotting.

Release of Monocytic Inflammatory Cytokines

The release of inflammatory cytokines (IL-1β, IL-6, IL-8 & TNFα) in the presence of

the alloy was assessed using a human acute monocytic leukemia cell line (THP-1 cells)

(ATCC, Manasass,VT). The cells were cultured in suspension in complete RPMI-1640

media (FisherScientific, Waltham,MA) for several days at 37 ̊C, 5% CO2 in a cell culture

incubator. The cells were then centrifuged and re-suspended at a concentration of 1 X

106 cells/ mL. A 1-mL cell suspension was placed in each well of a multiwell plate

containing 9.5mm diameter discs of Mg-Ca-Sr, S.S, and PLLA. Control cell population

Page 115: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

115

grown in plain growth media and media including lipopolysaccharide (LPS, 10 ng/mL)

were used as positive and negative controls for inflammatory response, respectively.

THP-1 cells were incubated in the presence of samples at 37̊C for 24 h under static

conditions. The cells were centrifuged and pelleted and the media supernatant was

collected and assayed. The concentrations of IL-1β, IL-6, IL-8 & TNFα in the

supernatants were determined using a multi analyte ELISA kit according to the

manufacturer’s instructions (Qiagen, Valencia, CA).

Statistical Analysis

All data in this study are reported as mean +/- standard deviations. An ordinary one

way ANOVA (Analysis of variance) with Dunnett’s multiple comparison post hoc

analysis was used to analyze and identify significant differences between the control

column and other columns. P-values <0.05 are reported as significant.

Results and Discussion

In spite of great advancement in the field, the major challenge while using

biodegradable metallic intracoronary stents is the prevention of thrombosis formation

and restenosis development after implantation108,110,232. This is primarily governed by

the interaction at the interface of the implanted biomaterial and the surrounding

biological environment. Therefore, the compatibility of the implanted stent material with

blood and blood components as well as surrounding vascular cells is of utmost

importance. In this study, the hemocompatibility and biocompatibility of a novel Mg-Ca-

Sr for its potential application as a vascular stent was investigated. Microstructure,

strength, and in vitro degradation behavior of this material and other extrusion

temperatures are reported in depth elsewhere. The microstructure exhibited large (50

µm x 10 µm) elongated grains interspersed with dynamically recrystallized grains

Page 116: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

116

around 1 µm in diameter, with a typical extrusion texture. The eutectic micro constituent

consisting of a combination of Mg, Mg2Ca, and Mg17Sr2 was broken up into strings of

particles in the extrusion direction. Mechanical properties of the material have been

investigated in the previous study. The alloy exhibited substantial mechanical properties

with yield strengths of 300 MPa and 250 MPa in tension and compression, respectively.

These mechanical properties are higher than that reported for other magnesium alloys

(WE43) as well as 316L annealed stainless steel which have been used in clinical

studies for vascular applications107,233. Additionally, the material exhibited a low in vitro

degradation rate in Hank’s solution, with hydrogen evolution ~0.022 mL H2/cm2/day.

Figure 5-1. Evaluation of cytotoxicity. Toxicity on HUVECs, expressed as a percentage of relative cytotoxicity, for three concentrations of alloy extract medium (10 %, 30 % & 50 %) in complete media. Data expressed as mean +/- SD, N=3. % Relative cytotoxicity of 10 % extract: 1.86 +/- 0.34 %, 30 % extract: 3.13 +/- 1.56 %, 50 % extract: 5.02 +/- 2.18 %, spontaneous TCP: 0 +/- 3.33 %. Data statistics based upon ordinary one-way analysis of variance (ANOVA) coupled with Dunnet’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

As a first step towards assessing the biocompatibility of this alloy cytotoxicity

response of the alloy was studied. Cellular response is one of the central factors

involved in selection, design and application of biomaterials. One of the rudimentary and

standardized techniques for assessing cellular response is by measuring in vitro

Page 117: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

117

cytotoxicity of the biomaterial, with appropriate cell types based on its eventual

application234. In this study the biocompatibility of the alloy was evaluated by assessing

cytotoxicity of alloy degradation products with human vascular endothelial cells. In this

study, vascular endothelial cells were chosen to closely mimic and model the cellular

environment that will be in direct contact with a vascular implant material. The cytotoxic

response elicited by the cells to the alloy was assessed via LDH release. HUVECs

exposed to 10, 30 and 50% diluted alloy degradation extracts were assessed for LDH

released into the media over a period of 24 h. The results as presented in Figure 5-1

revealed that the alloy extracts showed very low relative percent cytotoxicity (1.86+/-

0.34%, 3.13+/-1.56%, & 5.02 +/- 2.18 %) with all three extract concentrations (10, 30

and 50%) respectively. These results are consistent with previous reports on the

evaluation of cytotoxicity of this alloy composition with a pre osteoblast cell line (MC 3T3

E1) 222. The data further confirms the low cytotoxic response induced by the alloy on

human primary vascular endothelial cells which is an essential requirement of a

vascular stent material. Further the hemocompatibility of the alloy was assessed via

hemolysis, characterization of whole blood clotting behavior, platelet adhesion and

activation using circulating blood derived from domestic pigs. Hemolysis evaluation is

one of the most commonly used methods to determine preliminary hemocompatibility

and biocompatibility of a biomaterial235. The interaction of the biomaterial implant with

circulating blood can result in the release of hemoglobin due to RBC lysis. This process

can be influenced by chemical interaction (release of ions) of RBC with the degradation

products. The intactness of RBCs is important for maintenance of microcirculation. The

Page 118: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

118

lysed RBCs also release factors that can have detrimental effects such as platelet

activation and subsequent vasoconstriction236.

Figure 5-2. Hemolysis assessment. Percentage hemolysis of RBCs following incubation of diluted whole blood with Mg-Ca-Sr: 0.70 +/- 0.17 %, S.S: 0.02 +/- 0.05 %, PLLA: 0.16 +/- 0.28 %. Blood samples exposed to complete hemolysis with water (Max lysis: 100.00 +/- 9.23 %) and Blood samples with nothing added (No lysis: 0 +/- 0.11 %) were used to normalize the data (not shown in figure). Data expressed as mean +/- SD, N=9. Data statistics based upon ordinary one-way analysis of variance (ANOVA) coupled with Dunnet’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05). (C) Representative digital images of hemolysis by sample and controls. Hemolysis percentage correlated with absorbance of the supernatant.

Therefore, it is essential to determine the hemolytic percentage of the alloy for

vascular applications where the material is in direct contact with circulating blood. The

percentage hemolysis of the alloy as well S.S and PLLA controls as shown in Figure 5-2

indicate that the alloy caused very low and negligible (<1 %) hemolysis when incubated

with diluted blood for 2h. ISO 10993-4:2002 requires that the hemolysis rate of

hemocompatible materials be below 5 % 237. Previous reports assessing the

hemocompatibility of pure Mg, as well as, Mg-based alloys indicated that pure Mg as

well as most Mg based alloys were severely hemolytic. There are very few alloys such

A B

Page 119: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

119

as Mg–6Zn, Mg–1Si and WE43 that have been reported to show hemolytic rates less

than 5 % 219,233,238. High degradation rate of alloys have been known to result in drastic

pH variations which causes high hemolytic rates, therefore the sustained degradation

characteristics as exhibited by the alloy is beneficial to prevent excessive hemolysis233.

The results show that the Mg-Ca-Sr alloy causes very low hemolysis in comparison with

existing alloy systems thereby confirming the applicability of this alloy for vascular

applications.

Other than preliminary biocompatibility, the behavior of a potential stent material

with blood in terms of effect on clotting cascade is also an important criterion for

vascular application. Stent thrombosis caused by clot formation post implantation, leads

to catastrophic consequences that can result in death. Despite various anticoagulation

therapies after implantation, late stent thrombosis still remains a major challenge.108,208

Hence, one of the essential design considerations while choosing biomaterials for blood

contacting applications is the ability to resist thrombus formation. the thromboresistance

of this alloy along with other reference materials when in contact with activated porcine

whole blood was evaluated 229–231. The data (Figure 5-3) indicated that at 30 min, Mg-

Ca-Sr showed the lowest percentage clotting (P<0.05) when compared to S.S., PLLA

and Glass. At 60 min, even though there was some clot formation on the Mg-Ca-Sr

samples, the percentage clotting was still significantly lower than that initiated by S.S as

well as glass (P<0.05). The percentage clotting on PLLA samples however was

statistically similar to the alloy (P>0.05) at 60 min. Collectively, the data from the whole

blood clotting studies demonstrates that the Mg-Ca-Sr alloy exhibits significant

Page 120: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

120

thromboresistance which is essential for optimal performance of implants used for

vascular applications 71,109.

Figure 5-3. Whole Blood Clotting assessment. Percentage clotting of whole blood expressed as percentage decrease in absorbance calculated with respect to initial absorbance (A) at 30 min (Mg-Ca-Sr: 4.12 +/- 4.39 %, S.S: 34.08 +/- 5.31 %, PLLA : 25.22 +/- 10.78 % & Glass: 69.25 +/- 3.87 % ) (B) at 60 min (Mg-Ca-Sr: 43.46 +/- 11.79 %, S.S: 83.82 +/- 1.39 %,PLLA: 48.87 +/- 3.61 % & Glass: 84.82 +/- 1.84 % ). Data statistics based upon ordinary one-way analysis of variance (ANOVA) coupled with Dunnet’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05). (C) Representative digital images of clot formation on sample and control surfaces at 30 min and 60 min.

The ability to resist/delay clot formation is important to prevent thrombosis which

leads to fatal complications and subsequent failure of implant 84,109,216,229. It is important

A B

C

Page 121: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

121

to note that both Ca2+ ions and Sr2+ ions have been known to cause surface clot

formation, platelet adsorption and activation when exposed to blood plasma 225,239,240.

However, the Mg-Ca-Sr alloy shows desirable resistance to thrombus formation. This

effect is possibly due to the well-known anti-thrombotic effect of Mg ions 114,225,227,241.

Previous studies have shown that administration of Mg produces an inhibition of acute

stent thrombosis under high-shear flow conditions without any hemostatic or significant

hemodynamic complications113. Interestingly, many studies have also shown the ability

of Mg ions to act as natural Ca blockers and this may explain the improved blood

compatibility of the alloy despite the presence of clotting activating ions such as Ca 2+

and Sr 2+ ions 224,242. Further investigation is required to clearly understand the

mechanism of thromboresistance and synergistic interaction between the Mg, Ca and

Sr ions released by the alloy.

After confirming the thromboresistive behavior of the alloy, to further characterize

the interaction of this alloy with blood components, adhesion and activation platelets on

sample surface was quantified. Both platelet aggregation and activation on biomaterial

surfaces are major biomarkers of thrombogenicity of a biomaterial 75,243,244. Additionally,

platelets also play an important role in inflammation and immune responses. Platelets

release many factors that regulate inflammation which do not have any role in

hemostasis 245. Therefore, assessing the interaction of platelets on biomaterial surfaces

is vital for assessing its hemocompatibility as well as immune response 246. The

adhesion of platelets on the surface of the Mg-Ca-Sr alloy was quantified via release of

LDH by lysed platelets. The results as shown in Figure 5-4 A indicate that the number of

platelets adhered on the alloy surface was statistically similar to PLLA and S.S (P>

Page 122: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

122

0.05). However, the number of platelets adhered to the alloy was significantly lower than

collagen-coated plastic which was included as a control for positive thrombogenic

surface (P< 0.05). Although some surfaces may inhibit platelet adhesion, they can still

cause platelet activation 75

Figure 5-4. Platelet adhesion and activation. Data expressed as mean +/- SD, N=3. (A) Number of platelets adhered on Mg-Ca-Sr: 1.88E+06 +/- 4.93E+05, S.S: 2.30E+06 +/- 2.46E+05, PLLA: 9.69E+05 +/- 3.32E+05, Glass: 3.54E+06 +/- 4.46E+05 & Collagen coated plastic: 2.99E+06 +/- 8.34E+04 (B) Platelet activation by PF4 release in ng/ml by Mg-Ca-Sr: 5.33 +/- 1.02, S.S: 6.21 +/- 1.93, PLLA: 6.82 +/- 2.14, Glass: 5.88 +/- 1.45 & Collagen coated plastic: 25.66 +/- 2.40). Data statistics based upon ordinary one-way analysis of variance (ANOVA) coupled with Dunnet’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

When platelet activation occurs the platelets change shape and morphology

causing release of many intracellular granular contents245. Platelet factor 4 (PF4) is a

well characterized platelet-specific marker released from platelet alpha granules upon

activation247,248. Therefore, release of PF4 to measure the activation of platelets

contained within PRP when in contact with the samples in comparison with S.S and

PLLA controls was quantified as presented in Figure 5-4 B. The results indicate that the

amount of PF4 released by platelets in contact with the alloy was not statistically

A B

Page 123: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

123

different from PLLA or S.S (P> 0.05). However, the PF4 release caused by collagen-

coated plastic which was used a positive control for platelet activation in this study was

5-fold higher than that caused by the alloy (P<0.05). Some studies have reported the

increased platelet activation caused by metallic stents due to release of metal ions in

the blood plasma 104,249–251. Therefore, it was important to determine the effect of

platelet interaction with the alloy. The quantification of platelet adhesion and activation

indicated that the Mg-Ca-Sr alloy does not promote significant platelet adhesion or

activation, which are markers for both thrombus formation as well as inflammatory

response. This effect could potentially explained by the ability of Mg to reduce platelet

activation by inhibiting production of factors such as thromboxane A2252. Although

platelet aggregation and activation leads to inflammation, it is not the only mechanism of

inflammatory response. Therefore an assessment of inflammatory response was made

by assessing some markers of inflammation was made,since eliciting minimal

inflammatory response is also an important characteristic of a biomaterial 16,75. This

inflammatory response is mainly governed by interaction with monocytes and

macrophages 245,253. Monocytes have been known to mediate long-term chronic

inflammatory response. Peripheral blood monocytes when activated release many

cytokines and chemokines that play an important role in modulating inflammation

caused by the biomaterials254–256. Hence, to fully characterize the response to the Mg-

Ca-Sr alloy to evaluate its suitability as a vascular stent material, the release of pro-

inflammatory cytokines (IL-1β, IL-6 & TNF-α) and chemokines (IL-8) by THP-1 cells

which are widely studied to mimic monocytic response in cell culture models was

Page 124: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

124

quantified.253 The release of these factors was quantified via a custom designed ELISA

kit as described above (Figure 5-5).

Figure 5-5. Release of inflammatory cytokines/chemokines by THP-1 cells reported as absorbance at 450 nm. Data expressed as mean +/- SD, N=3. Blank absorbance value was not subtracted from the data sets for clarity. (A) Release of IL-1β, Mg-Ca-Sr: 0.03 +/- 0.01, S.S: 0.03 +/- 0.01, PLLA: 0.04 +/- 0.01, Untreated cells: 0.02 +/- 0.00 & LPS induced positive: 1.75+/- 0.16. (B) Release of IL-6, Mg-Ca-Sr: 0.02 +/- 0.01, S.S: 0.02 +/- 0.01, PLLA: 0.02 +/- 0.00, Untreated cells: 0.02 +/- 0.01 & LPS induced positive: 0.07+/- 0.00. (C) Release of IL-8, Mg-Ca-Sr: 0.47 +/- 0.06, S.S: 2.39 +/- 0.37, PLLA: 2.28 +/- 0.50, Untreated cells: 1.29 +/- 0.09 & LPS induced positive: 3.88+/- 0.06. (D) Release of TNF-alpha, Mg-Ca-Sr: 0.07 +/- 0.01, S.S: 0.13 +/- 0.01, PLLA: 0.13 +/- 0.01, Untreated cells: 0.11 +/- 0.00 & LPS induced positive: 0.51+/- 0.10. Data statistics based upon ordinary one-way analysis of variance. (ANOVA) coupled with Dunnet’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

THP-1 cells incubated with 10 ng/mL of Lipopolysaccharide (LPS) derived from

E. coli was included in these studies to induce an increased inflammatory response

while cells grown in culture media with no sample added was considered a negative

A B

C D

Page 125: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

125

control for inflammation 75,257. The data revealed that the alloy caused significantly lower

induction of inflammatory factors when compared to LPS induced controls.

The results also showed that the release of IL-1β and IL-6 (Figure 5-5A & B) caused by

the alloy was statistically similar to that induced by S.S, PLLA as well the negative

control (P>0.05). Interestingly, the quantification of release of IL-8 and TNF-alpha

(Figure 5-5 C & D) by alloy showed significantly lower amounts when compared to S.S,

PLLA and negative control (P<0.05). This data corroborates the findings obtained from

platelet studies and confirms that the alloy elicits very low inflammatory response.

Although a more thorough investigation of alternate and synergistic inflammatory

pathways is required to completely understand the inflammatory response to the alloy,

the current data suggests preliminarily confirms the minimal inflammatory response

invoked by the Mg-Ca-Sr alloy.

Summary

Structural and architectural cues presented to cell systems via biomaterials elicit

important cellular responses in them including alteration of cell shape, alignment,

differentiation, growth and tissue formation14,258. The response between a biomaterial

and cell system can be varied across multiple length scales. The capability of cell

systems to sense microarchitectural, physical as well mechanical properties of the

substrate underneath falls under the meso scale (10 to over 100 µm). This capability is

made use of while designing biomaterials to modulate specific cellular response for a

variety of applications. Cardiovascular biomaterials can be designed to alleviate

complications arising from different cardiovascular diseases, such as atherosclerosis,

myocardial infarction, peripheral artery disease etc. Chapters 4 and 5 covered two

important classes of cardiovascular biomaterial applications, namely, small diameter

Page 126: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

126

vascular grafts and vascular stents, respectively. This work was focused on studying the

interaction of these biomaterials with in vitro models of the vascular environment to

understand and develop strategies to repair damaged vasculature and enable

regeneration.

The first part of the study was focused on development of a novel bilayered,

construct for application as a vascular graft. The graft was designed to mimic the

biological and architectural aspects of native blood vessels. The design consisted of two

layers, a solid luminal layer made of elastomeric, biodegradable POC and a fibrous

medial layer consisting of a mixture of collagen, POC and elastin. The rationale behind

the design is to provide a solid lumen for the formation of a monolayer of endothelial

cells and fibrous medial layer enriched with ECM proteins to enable formation of an

interconnected network of SMCs. The individual layers of the graft were separately

fabricated and assessed for cell attachment and proliferation. The constructs

mechanical properties were also assessed. Overall the results showed that the graft

was biocompatible and was able to support attachment and growth of vascular cells.

More interestingly, the mechanical properties evaluation revealed that the compliance of

the fabricated graft matched native artery values. This property is very important to

prevent implant failure due to mismatch in mechanical properties195,203,204. This graft is

currently being developed to recruit circulating stem cells to promote integration into

vasculature.

The second part of the study evaluated the hemocompatibility of an Mg-Ca-Sr

alloy to determine its utility as a candidate material for vascular applications. Overall,

this study showed that the alloy is biocompatible and invokes very low cytotoxic

Page 127: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

127

response when cultured with a mature vascular endothelial cell population. The alloy

sample was hemocompatible and exhibited excellent thromboresistance as indicated by

low hemolytic percentage as well as delayed and reduced clotting when in contact with

whole blood. The alloy also did not cause significant platelet adhesion and activation.

Finally, it also does not induce significant release of pro-inflammatory factors confirming

minimal inflammatory response. This data collectively confirms the suitability of the Mg-

Ca-Sr alloy for potential application as a vascular stent material. The studies combined

investigated the effect of biomaterial structural cues on the meso scale on their ability to

be used in vascular environments. The studies involved evaluating some important

characteristics of these materials in terms of their biocompatibility, hemocompatibility,

ability to support vascular cell types, mechanical and biochemical behavior. These

factors are essential in preventing vascular damage due to complications such as

thrombosis, toxicity as well as, fostering healthy remodeling and regeneration of

vasculature. Combined, these strategies are useful for developing suitable biomaterials

for vascular engineering.

Page 128: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

128

CHAPTER 6 MACROSCALE SYSTEMIC CUES- PART 1- EFFECT OF SIMULATED

MICROGRAVITY ON DIFFERENTIATION AND DOWNSTREAM FUNCTION OF PORCINE BLOOD DERIVED ENDOTHELIAL PROGENITOR CELLS

Introduction

Mammalian cells, especially stem cells have been known to be highly sensitive to

environmental cues that are provided to them128,259,260. These cues can be a variety of

different factors such as pH, temperature, pressure, nutrient supply and waste removal,

mechanical, electrical and magnetic stimuli, etc. There is a significant amount of

evidence in literature suggesting that physical stimuli may affect gene expression and

significantly increase the biosynthetic activity in a range of different cells176,261,262. The

ability of physical stimuli to modulate favorable cell response has caused the

development of functional simulations systems to create models for tissue engineering

in vitro. Besides providing tissue engineering solutions, these studies are also important

to understand the effect of mechanotransduction on cellular systems.

This is important especially in vascular tissue engineering since the endothelium

has been shown to respond to physiological mechanical cues, such as shear stress,

cyclic stretch, and alterations to gravity which cause them to undergo significant

morphological and functional changes129,143,260,263,264. One such specialized macro

environmental model is microgravity in space flight, which provides a unique opportunity

to study the effect of gravitational changes on cells.

Within the past 50 years of human space flight missions, several physiological

threats to astronauts have been identified. These include: losses in bone mineral

density, muscle atrophy, shifts in the distribution of body fluids, and cardiovascular

deconditioning.135,265,266 In general, these clinical manifestations are a result of what is

Page 129: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

129

termed as cardiovascular deconditioning or the inability of the cardiovascular system to

function properly. There has been a significant amount of research focused on studying

the negative effects of cardiovascular degeneration and the development of possible

strategies and countermeasures to address them267,268.

Proper function of the cardiovascular system is largely dependent on the

maintenance of healthy functioning vascular cells, namely endothelial cells that line the

vasculature and are in direct contact with circulating blood169,269. The endothelial cells

(ECs) that line the vasculature form a thin layer called the endothelium, and this layer of

the vasculature plays a critical role in regulating blood flow, vascular permeability, and

maintaining an actively anti-thrombogenic surface. The ability of the endothelium to

perform these functions is largely governed by their highly regulated secretory

capabilities270,271.

EC dysfunction has been implicated as an important factor in weightlessness-

induced cardiovascular deconditioning143. In addition to resident ECs, circulating

progenitor cells recruited from the bone marrow into circulation are also involved in

maintaining vascular homeostasis272,273 . While great attention has been placed on

understanding the effects of weightlessness on the endothelium, much less work has

been done to study the effect of microgravity on the “repair cells”, circulating endothelial

progenitor cells (EPCs).These EPCs reside in the bone marrow and are released into

circulation upon stimulation effected by specific triggers270,273–275. Once in circulation

they can integrate into the vasculature, differentiate into endothelial cells, and

participate in the repair of damaged vascular tissues. Recent work has shown that the

amount and function of EPCs are both significantly impaired due to the incidence of

Page 130: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

130

cardiovascular diseases. Interestingly, the number as well as the health of circulating

EPCs have been identified to be indicators of the onset as well as the progression of

cardiovascular diseases276–278. Additionally, due to their natural ability to act as ‘repair’

cells for the cardiovascular system, several in vitro studies and subsequent animal

studies have exploited their therapeutic potential279,280. Recently these cells were also

used in clinical studies with human subjects to alleviate different cardiovascular

conditions with promising results281.

A common feature of the effects of altered gravitational conditions on different

cellular systems is that the affected tissues rely on tissue-specific stem cells for

regeneration and repair. Understanding and replicating the behavior in such a condition

such as altered microgravity can lead to development of stem cell based therapy for

cardiovascular degeneration on earth. A recent study by Chiu et al. has shown that

human cord blood stem cells when cultured under simulated microgravity proliferated

and trans-differentiated into 3D structures resembling vascular tubules while exhibiting

vascular endothelial phenotype expressions140. Many such studies on the effect of stem

cells under alternate culture environments especially simulated microgravity is currently

being studied to develop regenerative strategies. Current research is also studying the

hypothesis that cardiovascular deconditioning due to space travel may potentially trigger

faster maturation of stem cells and therefore enhancing their regenerative potential.

The objective of this study is to fill the gap in knowledge regarding the effects of

microgravity on the downstream function of EPCs to understand their role in

cardiovascular repair during space travel and to propose a stem cell based therapeutic

strategy to counteract the negative effects and regenerate vascular tissue. More

Page 131: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

131

specifically this work is targeted towards studying the effect of altered gravity conditions

on the differentiation of EPCs to ECs and the downstream functionality of the ECs

themselves.

Towards this goal, this study was conducted in two parts, the first part focused

on understanding the effect of simulated microgravity on the differentiation and function

of EPCs after being pre-exposed to simulated microgravity while the second part

involved investigating of differentiated EPCs when cultured under simulated

microgravity conditions. These two parts will be covered in Chapters 6 and 7

respectively. With these goals in mind, the following specific aim was established and

studied in this work.

Specific Aim 3

Study and understand the effect of simulated microgravity on the function of

vascular stem cells to assess their potential to be used as a therapeutic strategy for

vascular repair and regeneration.

Before the methodology and results of the ensuing study can be discussed, it is

important to establish a definition and understanding of simulated microgravity and the

bioreactor culture set up that was used to generate this condition. Microgravity refers to

the condition where the effect of gravitational pull on objects seems to be diminished in

space265. Although the golden standard to understand effect of this condition on cell

systems is by studying their behavior in real space travel, spaceflight missions are often

very rare and expensive. Therefore, for preliminary studies, researchers have

developed ground-based models that can simulate this condition on Earth to prepare

their future space missions. Many strategies and devices with considerable success in

simulating microgravity have been developed for tissue engineering purposed119.

Page 132: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

132

In this study a NASA designed, rotating wall vessel (RWV) bioreactor that mimics

certain functional aspects of microgravity on earth (Synthecon, Houston, TX) was

utilized. The RWV was designed and developed at NASA’s Johnson Space Center to

simulate the effects of microgravity on cells in a ground-based culture system134,282.The

bioreactor set-up consists of a cylindrical vessel/chamber that rotates around its

horizontal axis with a coaxial tubular silicon membrane for gas exchange. (as shown in

figure below) 119,126,127,129

Figure 6-1. RWV bioreactor set-up (A) Digital image of Synthecon Slow Turning Lateral Vessel with rotor base, (B) RWV bioreactor detachable chamber.

To study the effect of this simulated microgravity on EPCs a porcine model was

chosen since it is widely preferred as a model system for vascular research due to its

physiological and hemodynamic similarities to the human vascular system283.

Additionally, a reproducible and consistent method to induce differentiation of porcine

blood derived EPCs has been reported85 . Lastly, the effect of altered gravity culture

conditions on the function of mature porcine endothelial cells has been studied in the

past284. However, the effects of short term hypogravity or microgravity exposure, on

circulating progenitor cells have not been reported before. This chapter describes the

Page 133: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

133

effect of simulated microgravity on the differentiation of circulating progenitor cells

derived from pig blood along an endothelial pathway. This work also describes the

effect of short term exposure to simulated microgravity, on the downstream functionality

of the differentiated endothelial cells such as release of soluble factors, prostaglandin

E2 (PGE2) and nitric oxide (NO), proliferation, migration and ability to regulate clotting

mechanisms. This study is the first step towards understanding the functionality of

EPCs in altered condition to assess their ability to be used therapeutically to repair and

regenerate vasculature.

Methods and Materials

Isolation of Peripheral Blood Mononuclear Cells from Porcine Blood

About 50 ml of circulating peripheral blood was collected from the exterior jugular

vein of domestic pigs (Department of Animal Sciences, University of Florida) in the

presence of acid citrate dextrose anticoagulant. All blood collection procedures were

performed in accordance with the rules and regulations of the Institutional Animal Care

and Use committee at the University of Florida. Within 4h of blood collection, the

peripheral blood mononuclear cells (PBMNCs) were isolated via density gradient

centrifugation as previously described 85,270. An average density of 140-160 million

mononuclear cells obtained from the isolation process were suspended in endothelial

cell growth media (EGM-2) (Lonza, Walkersville, MD) supplemented with 20% fetal

bovine serum. Porcine aortic endothelial cells (PAECs) (Cell Applications, SanDiego,

CA) between passages 4-6 cultured in the EGM-2 served as a positive control for

mature EC function and response.

Page 134: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

134

Rotating Wall Vessel Bioreactor System

The culture of cell systems in actual microgravity can be achieved only through

parabolic flights, rockets, space crafts, or laboratories available on the International

Space Station (ISS). However, the ability to culture various cell systems in real

microgravity is limited by the cost involved in performing these experiments on space

missions. Therefore a variety of devices have been designed to mimic some aspects of

real microgravity on earth 119. In this study, some aspects of simulated microgravity (µg)

conditions were established using a RWV bioreactor consisting of a rotor base and

detachable cell culture chambers (Figure 6-1 A). The chamber consists of a silicone

membrane for gas exchange as shown in Figure 6-1 B. Using this set up, the time-

averaged gravitational vector acting on these cellular assemblies is reduced to about

10−2 g. For simplicity, this condition is referred to as simulated microgravity. The validity

of the RWV model to simulate certain aspects of weightlessness has been verified and

reported extensively in literature 119,127,136. Additionally, the use of RWV to simulate

aspects of microgravity has been confirmed and validated by experiments in real

microgravity in the past 133,135,285,286.

Microgravity Culture of PBMNCs

The bioreactor set up as described above was placed in a cell culture incubator

maintained at 37°C and 5% carbon dioxide. The experimental set-up included 1)

PBMNCs suspended (18x106 cells in 50ml of EGM2) inside the bioreactor chamber for

microgravity exposure; and 2) PBMNCs seeded at a density of 2-3x105 cells/cm2 onto

fibronectin coated tissue culture polystyrene (TCP) multiwell plates (BD Biosciences,

Franklin Lakes, NJ) to be cultured statically with no simulated microgravity exposure. To

maintain conditions of microgravity the bioreactor chambers containing PBMNCs were

Page 135: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

135

rotated at a minimum speed of 5rpm. Following 6, 12 or 18h-µg exposure, the PBMNCs

were collected, then seeded onto fibronectin coated TCP plates and cultured statically

for 7-10 days.

Differentiation, Colony Formation and Immunocytochemistry

The statically cultured cells, both those pre-exposed to microgravity and those in

normal gravity for the entirety, were visually assessed to observe colony formation

every day for 8 days using an inverted phase-contrast microscope (Nikon TE2000U,

Melville, NY). The first appearance of densely packed, isolated, cobblestone

morphology, endothelial-like cells in each field of view was recorded as indicative of EC

differentiation. The number of colonies and the rate of new colony formation was

recorded.

PE-like cells were examined using immunohistochemical analysis to confirm

endothelial cell phenotype. Briefly, cells were fixed in 4% paraformaldehyde,

permeabilized, if required, using 0.2% Tween-20 and then blocked using 3% bovine

serum albumin. Following which, the cells were incubated overnight at 4⁰C with a

primary antibody (diluted 1:100) against endothelial nitric oxide synthase (Abcam,

Cambridge, UK) and CD31 (diluted 1:100) (AbDSerotec, Raleigh, NC). Subsequently,

the probed cells were incubated with a fluorescein isothiocyanate tagged isotype-

matched secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA) and nuclei

were stained using Hoechst 33342 (ThermoScientific, Waltham, MA). Fluorescent

images were captured using an inverted fluorescent microscope (Nikon TE2000U).

Endothelial-Like Cell Migration and Proliferation

Cell migration was assessed using the established scratch assay 287. Briefly, PE-

like cells differentiated after 6, 12, and 18h of pre-exposure to simulated microgravity as

Page 136: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

136

well as those cells differentiated in normal gravity, were cultured to approximately 90%

confluence on multi-well TCP plates in the culture media as described above. A linear

scratch was made through the monolayer using a pipette tip to create a “cleared” area,

then the cells were incubated at 37° C for 18h to allow for migration. Digital images of

the scratch surface were taken before and after incubation and the percentage area

covered by the migrating cells was quantified using ImageJ software version 1.46.

Cell proliferation was assessed via changes in DNA concentration from cultured

cells over time, quantified using Quant-it picogreen dsDNA assay kit (ThermoScientific).

Briefly, PE-like cells differentiated after 6, 12, and 18h of pre-exposure to microgravity

as well as normal gravity cultured controls, were seeded on 12-well TCP plates at a

seeding density of 2500 cells/cm2. At 24hr increments over 4 days, the cells were lysed

to collect and quantify the intracellular DNA. The concentration of DNA contained within

the lysates were normalized by subtracting the initial seeded DNA concentration from

the concentration at each time point for each sample.

Nitric Oxide and Prostaglandin Secretion

NO and PGE2 secretion was evaluated in PE-like cells differentiated after 6, 12,

and 18h of pre-exposure to simulated microgravity as well as the cells differentiated

under normal gravity. The amount of total NO and PGE2 secreted by the cells into the

culture medium over a period of 24h for each cell population was determined using the

total NO assay kit and Prostaglandin E2 Assay kit (R&D systems, Minneapolis, MN)

following manufacturer protocols. Briefly, the concentration of NO measured is based

on the enzymatic conversion of nitrates to nitrites and the detection of total nitrite

concentration via the Greiss reaction. PGE2 concentration was assessed via enzyme

Page 137: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

137

linked immunosorbent assay. The concentration of both NO and PGE2 was normalized

to cell number and expressed in terms of concentration per cell.

Clotting Kinetics

Following previously reported methods, the kinetics of both plasma and whole

blood clot formation was assessed 85,270. Briefly, plasma and whole blood were exposed

to monolayers of PE-like cells differentiated after 6, 12, and 18h of pre-exposure to

simulated microgravity as well as cells differentiated under normal gravity, and the

kinetics of clot formation were reported. In this study, PAEC served as a positive control

for anti-thrombogenic endothelial cell function, and bare TCP serves as a negative

control representing the kinetics of uninhibited plasma and blood clotting.

Statistical Analysis

All data are reported as means +/- standard deviations. Numerical means were

compared using a student 2-sample t-test. Analysis of variance (ANOVA) with Newman-

Keuls multiple comparison test post hoc analysis was employed to identify significant

differences between 3 or more means. P-values <0.05 was reported as significant.

Results and Discussion

This study reports the effect of early exposure to simulated microgravity on the

differentiation and downstream function of porcine blood derived progenitor cells.

Although multiple studies have investigated and reported the effect of altered gravity

conditions on the behavior of adult endothelial cells, there are limited reports on its

effect on circulating progenitor cells143. Since these cells have been shown to be

involved in vascular repair mechanisms in the body, it is important to understand the

functionality of these cells when exposed to short term microgravity, to better

understand effects of cardiovascular repair in space and develop stem cell based

Page 138: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

138

therapies to repair, preserve and regenerate vasculature. Previous studies have

reported significant alterations to morphological characteristics and changes to gene

and protein expression in mature endothelial cells within a few minutes to a few days of

microgravity exposure143,288–290. Moreover, the changes in expression of immunological

cytokines in PBMNCs and the recovery of their functions on short term exposure to

simulated microgravity ranging from within a few hours to up to 3 days has been

reported before291.

In this study, circulating progenitor cells contained within the PBMNC fraction of

porcine whole blood were exposed to simulated microgravity, using a RWV bioreactor

designed originally by NASA to simulate reduction in earth’s gravity (1g) to micro-scale

(µg) on cultured cells. The time-averaged gravitational vector acting on these cells is

reduced to about 10-2 g using this set-up. After exposing the circulating progenitor cells

to 6, 12 and 18h of microgravity, the cells were then cultured on fibronectin coated TCP

under static conditions and their ability to differentiate into endothelial cells was

assessed by the formation of colonies with EC specific ‘cobble-stone’ morphology.

(Figure 6-2A) In all cases the observation for colonies was concluded after 8 days after

which it was difficult to discern discrete isolated colonies. Cells exposed to microgravity

for longer than 18h showed significantly reduced or no colony formation when cultured

on fibronectin coated TCP. This indicated the existence of a possible threshold time of

microgravity exposure beyond which the differentiation ability of these PBMNCs may be

permanently impaired. On assessing the kinetics of EC colony formation, it was

observed that the average rate of endothelial cell colony formation decreased with

increased time of exposure to microgravity between 3-6 days of culture (Figure 6-2 B).

Page 139: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

139

While the normal gravity showed the highest rate of colony formation as indicated by an

average of 10 colonies/day.

Figure 6-2. Differentiation and characterization of PE-like cells. Representative phase contrast image of PBMNC derived endothelial colony formation after 4 days of static culture (4x), Scale bar 200 µm. The region indicated by the dotted circle shows a newly differentiated PE-like cell colony (B) Graph showing the kinetics of colony formation of microgravity exposed PE-like cells vs cells maintained in normal gravity. (0h µg: 10 colonies/day, 6h and 12h µg: 8 and 6 colonies/day vs 18h µg: 2.67 colonies/day, respectively) (C) Representative phase contrast image of characteristic endothelial ‘cobble-stone’ morphology of PE-like cells (20X). Representative 20X fluorescent images of positive staining of both 12h µg exposed as well as normal gravity exposed PE-like cells for (D-E) endothelial nitric oxide synthase (F-G) CD31 shown in green, nuclei were counterstained with Hoechst 33342 shown in blue. Scale bar is 100 µm.

The control population of PBMNC, cultured under normal gravity conditions

formed colonies at the earliest period of 3 days compared to microgravity exposed

Page 140: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

140

populations which did not form any colonies as early as the third day of culture. The

endothelial phenotype of both the microgravity exposed and normal gravity cultured

cells were confirmed by their characteristic cobble stone morphology (Figure 6-2 C) and

positive staining for eNOS (Figure 6-2D-E) and CD31 (Figure 6-2 F-G).

In the past, the decrease/delay in colony formation of ECs from EPCs have been

associated to pre-existing cardiovascular complications292,293.This delay in differentiation

indicates a possible detrimental effect of simulated microgravity on the onset of

differentiation of EPCs to ECs that may affect the regulation of cardiovascular health by

these cells. Also, the ability of simulated microgravity to inhibit bone marrow

hematopoetic stem cell differentiation has been confirmed by Plett et al. (2004)294.

Further, the effect of this altered condition on the downstream functional abilities

of the differentiated endothelial-like cells that were pre-exposed to microgravity was

investigated. The differentiated cells were assessed for their proliferation and migratory

ability. Migration was assessed by observing and quantifying the migration of cells into

an area cleared of cells (Figure 6-3 A). The quantification of percentage area of the

scratch covered by the migrating cells (Figure 6-3 B), showed that the 6h-µg exposed

and normal gravity cultured PE-like cells migrated the most followed by 12h-µg exposed

cells (P<0.05). Finally, the 18h-µg exposed PE-like cells migrated the least (P<0.05),

indicating a direct decrease in migration ability of the EC with increased time of

exposure of the progenitor cells under simulated microgravity.

Page 141: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

141

Figure 6-3. Proliferative and migratory functions of simulated microgravity pre-exposed PE-like cells (A) Representative phase contrast images showing migration of microgravity exposed PE-like cells over 18 h into a scratch made in the cell monolayer vs the migratory response of cells maintained in normal gravity. Representative results showing (B) Quantification of % area covered by migration of cells into the scratch. Data expressed as mean +/- SD, N=6; (6h µg exposed cells: ~91+/-8 %; 0h µg ~93+/-10%; 12h µg: ~78+/- 4%; 18h µg: ~38+/-3%) (C) Proliferation kinetics of microgravity exposed PE-like cells vs cells maintained in normal gravity as measured by DNA concentration in cell lysates. (Average initial rate of proliferation reported as rate change in concentration of DNA content (pg/day), 6h µg exposed cells: 2.64E+05 +/- 8.76E+03; 12 µg: 3.01E+05 +/- 2.74E+04, 18 µg: 4.98E+05 +/- 1.49E+03, 0h µg: 6.93E+05 +/-9.40E+02). Data expressed as mean +/- SD, N=3. Data statistics based upon one-way analysis of variance (ANOVA) coupled with Tukey’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

The effect of pre-exposure to µg on the proliferative capacity of the EC-like cells

was also assessed by quantifying the change in DNA over time in cultured cells. The

data show that 6 and 12h of pre-exposure to µg altered the initial rate of proliferation

most significantly. (Figure 6-3C). In contrast, the PE-like cells pre-exposed to

Page 142: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

142

microgravity for 18h proliferated faster than both (Figure 6-4C, P<0.05). As expected,

the PE like cells differentiated under normal gravity showed no inhibition in proliferation,

but rather showed the greatest amount of proliferation (P<0.05).

These data collectively show that both the migratory ability and proliferative

ability of ECs are impaired by pre-exposure of the EPCs to microgravity. While

increased exposure time to µg impaired the migration of the microgravity exposed cells,

the proliferation of these cells seems to be enhanced by increased time of microgravity

pre-exposure. This indicates a possible recovery of the proliferative ability of the PE-like

cells on longer times of exposure of the EPCs to simulated microgravity. A similar

recovery of immune functions of human PBMNCs after their short term exposure to

simulated microgravity has been reported291. Further investigation of this recovery

mechanism may result in the development of possible countermeasures for treating

cardiovascular deconditioning to enhance recovery291,295,296.

A healthy endothelium can control and regulate thrombogenic properties and

clotting cascades in the vasculature by releasing anti-thrombogenic factors into the

blood stream. The ability of porcine EPC derived ECs to inhibit clotting mechanisms has

been shown before85,270,271,297. Hence it is important to assess the ability of ECs derived

from microgravity pre-exposed progenitors to regulate clotting mechanisms. To evaluate

the anti-thrombogenicity of these cells, their ability to release soluble factors such as

NO and PGE2 into the surrounding medium was first investigated. Prostacyclin and

Prostaglandin E2 have been known to be released from the endothelium after

stimulation by vasoactive agents and thrombin243. Prostacyclin is predominantly

released by the endothelium of large diameter blood vessels, while the ECs from

Page 143: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

143

smaller diameter blood vessels release predominantly prostaglandin E2298. Both of

these factors contribute to platelet inactivation and regulate clotting mechanisms243.

Another important molecule that regulates clotting mechanisms in the body is nitric

oxide, which is produced by endothelial nitric oxide synthase (eNOS)243,299. Both these

molecules synergistically regulate anti thrombogenicity in the body. The results show a

decrease in secretion of soluble PGE2 by 6h-µg exposed cells (P<0.05) while there

was no significant difference in the levels of PGE2 between the other microgravity

exposed cells as well as the control cells cultured under normal gravity (Figure 6-4 A).

Figure 6-4. Representative results of quantification of release of soluble factors. (A) Prostaglandin E2 over 24 h by microgravity exposed PE-like cells vs cells maintained in normal gravity (0h µg). (Data shown are Mean +/- SD; n=4). (PGE2 concentration in pg/ml/cell; 6h µg: 4.06E-06 +/- 1.71E-06, 12h µg: 1.86E-05 +/- 4.8E-06; 18h µg: 1.84E-05 +/- 4.84E-06; 0h µg: 1.34E-05 +/- 3.33E-06) (B) Nitric Oxide over 24 h by microgravity exposed PE-like cells vs cells maintained in normal gravity (0h µg). (NO concentration; in µmol/L/cell, 6h µg: 1.66E-06+/- 7.12E-07, 12h µg: 3.135E-05 +/- 3.29E-06; 18h µg: 2.88E-05 +/- 2.89E-06; 0h µg: 2.75E-05 +/- 6.35E-06). Data shown are Mean +/- SD; n=3. Data statistics based upon one-way analysis of variance (ANOVA) coupled with Tukey’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

The assessment of release of soluble NO indicated a similar trend where the

amount of NO released by 6h-µg exposed cells was significantly lower (P<0.05) when

Page 144: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

144

compared to other microgravity exposed cells as well as normal gravity cultured cells

(Figure 6-4B).

This data shows the ability of short term microgravity exposure of EPCs to inhibit

the regulation of release anti thrombogenic molecules into the surrounding medium.

However, with increased time of microgravity exposure, a potential recovery effect is

also observed where the 12h and 18h-µg exposed cells secrete NO and PGE2 in levels

comparable to the cells cultured under normal gravity. Previous reports have shown an

increase in the release of both the factors by mature ECs when cultured under

microgravity conditions 143. However, the effect of short term microgravity exposure of

progenitor cells, to alter the release of soluble factors by ECs derived from them have

not been reported before. This data is in coherence with whole blood clotting and

recalcified plasma clotting studies subsequently performed on these cells. Plasma

recalcification clotting profiles indicated that as expected the TCP surface induced

highest rate of plasma clotting within the first 90 seconds followed by 6h µg exposed

PE-like cells (Figure 6-5 A, P<0.05). Following the clotting rate for 6h-µg exposed cells

are the clotting times for the 12h and 18h-µg exposed cells. Normal gravity cultured

cells clotted the next while the last to clot was the plasma exposed to the PAEC control

cells (P<0.05). Similarly, the whole blood clotting kinetics showed that after 20 min the

average rate of whole blood clot formation was the highest for TCP surface (Figure 6-5

B, P<0.05) followed by whole blood exposed to 6h, 12h and 18h-µg exposed cells,

whose average clotting rates were lower than TCP control (Figure 6-B). While the whole

blood exposed to PAEC and cells cultured under normal gravity showed the greatest

inhibition of clotting.

Page 145: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

145

Figure 6-5. Representative result showing quantification of blood clotting studies. (A) recalcified plasma clotting, rate of plasma clotting expressed as change in absorbance/min, TCP: 0.537 +/- 1.26E-03, 6h µg: 0.510 +/- 1.25E-03, 12h µg: 0.368 +/- 2.5E-03, 18h µg: 0.3635 +/- 1.5E-03, 0h µg: 0.335 +/- 1.02E-03, PAEC: 0.1567 +/- 2.14E-03 (B) whole blood clot formed over time on monolayers of microgravity exposed PE-like cells vs cells maintained in normal gravity, PAECs and TCP. Error bars have been omitted for visual clarity. Average rate of clotting in g/min on TCP: 2.733E-03 +/- 4.42E-04; 6h µg cells: 2.08E-03 +/- 1.71E-04; 12h µg: 1.89E-03 +/- 4.32E-04; 18h µg: 1.633E-03 +/- 3.93E-05; PAEC: 1.175E-03 +/- 1.37E-04 and 0h µg: 1.288E-03 +/- 2.92E-04. Data statistics based upon one-way analysis of variance (ANOVA) coupled with Tukey’s multiple comparison test, N.S. indicates no significant difference (P > 0.05) and * indicates a statistically significant increase (P<0.05).

This data collectively shows that exposure to microgravity in the early progenitor

stages negatively influences the anti-thrombogenic function of PE-like cell relative to

those differentiated under normal gravity. The data shows that within the 3 time points

of µg exposure, the longer exposure times showed greater inhibition of clotting, which

may be attributed to the increased ability to release soluble anti-thrombogenic factors,

NO and PGE2. Although previous reports have shown the ability of simulated

microgravity to directly affect platelet functions 144, to the knowledge, the effect of

simulated microgravity on anti-thrombogenic properties of ECs, such as inhibition of

blood and plasma clotting has not been reported so far. This work provides an insight

into the microgravity mediated alterations to regulation of clotting mechanisms by

Page 146: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

146

circulating stem cell derived endothelial cells which is important to assess their role in

cardiovascular regeneration during space travel. As an extension of this study, Chapter

7 deals with the functionality of blood derived endothelial cells when cultured under

simulated microgravity after differentiation.

Page 147: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

147

CHAPTER 7 MACROSCALE SYSTEMIC CUES- PART 2- EFFECT OF SIMULATED

MICROGRAVITY ON FUNCTION OF HUMAN BLOOD DERIVED ENDOTHELIAL CELLS

Introduction

Within the past 50 years of human space flight missions, several physiological

threats to astronauts have been identified. These include: losses in bone mineral

density, muscle atrophy, shifts in the distribution of body fluids, and cardiovascular

deconditioning135,265,266. There has been a significant amount of research focused on

studying the negative effects of cardiovascular degeneration and the development of

possible strategies and countermeasures to address them267,268. Proper function of the

cardiovascular system is largely dependent on the maintenance of healthy functioning

vascular cells, namely endothelial cells that line the vasculature and are in direct contact

with circulating blood169,269. The endothelial cells (ECs) that line the vasculature form a

thin layer called the endothelium, and this layer of the vasculature plays a critical role in

regulating blood flow, vascular permeability, and maintaining an actively anti-

thrombogenic surface. The ability of the endothelium to perform these functions is

largely governed by their highly regulated secretory capabilities270,271. The endothelium

has also been shown to respond to physiological mechanical cues, such as shear

stress, cyclic stretch, and alterations to gravity which cause them to undergo significant

morphological and functional changes129,143,260,263,264.

Supplementary to ECs, circulating progenitor cells (CPCs) also play a role in

regulating the normal functioning of the vasculature. These CPCs reside in the bone

marrow and are released into circulation upon stimulation270,273–275. Once in circulation

they can integrate into the vasculature, differentiate into endothelial cells, and

Page 148: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

148

participate in the repair of damaged vascular tissues. The long-term goal of this study is

to fill the gap in knowledge regarding the effects of microgravity on the downstream

function of CPCs after they have differentiated into ECs. These stem cells serve as a

source of healthy vascular cells and play a very important role in the maintenance of

vascular homeostasis271,272,277,279. While the effect of weightlessness on mature ECs

has been studied extensively, there is a dearth of information regarding the effect of

microgravity on ECs derived from the circulating stem/progenitor cell population. Hence,

it is important to study the effect of simulated microgravity on the functions of these cells

to understand their implications during space flight and their possible role in the onset of

cardiovascular dysfunction.

As a first step towards establishing an understanding of the effect of simulated

microgravity on circulating progenitor derived ECs, modeled microgravity was studied

using a ground based rotating wall vessel (RWV) bioreactor. The RWV was designed

and developed at NASA’s Johnson Space Center to simulate the effects of microgravity

on cells in a ground-based culture system134,282. The bioreactor set-up used in this study

consisted of a cylindrical vessel/chamber that rotates around its horizontal axis with a

coaxial tubular silicon membrane for gas exchange 119,126,127,129. The results obtained

using simulated microgravity conditions utilizing the RWV have been confirmed and

validated by experimental models in space flights to outer space133–135,285.

This study reports the effect of simulated microgravity on the downstream

functions of human blood derived ECs such as: proliferation, release of soluble factors

nitric oxide (NO), interleukin-6 (IL-6), tissue plasminogen activator (tPA), gene

expression of HSPA4 (gene encoding heat shock protein 70 kDa A) and anti-

Page 149: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

149

thrombogenic potential. More specifically this work is focused on the ability of these

cells to adapt to hypergravity conditions and modulate these functions after short term

culture and expansion under simulated microgravity conditions. To the knowledge, this

is the first report that provides valuable insight into the functionality of human blood

progenitor cell derived ECs following exposure to simulated microgravity. This work

provides a foundation for future work to elucidate vascular repair mechanisms taking

place in the body in outer space.

Materials and Methods

Isolation of Peripheral Blood Mononuclear Cells from Human Blood

About 45 ml of circulating peripheral blood was collected from healthy donors by

venipuncture in the presence of heparin (158 USP) anticoagulant in vacutainer tubes

(Thermo Fisher Scientific, Waltham, MA). For clotting studies alone, blood was collected

in vacutainer tubes containing Acid Citrate Dextrose (ACD) solution (Thermo Fisher

Scientific) as the anti-coagulant. All blood collection procedures were performed in

accordance with the rules and regulations of the Institutional Review Board at the

University of Florida. Within 2h of blood collection, the peripheral blood mononuclear

cells (PBMNCs) were isolated using density gradient centrifugation as described in

detail elsewhere85,270. Approximately 80 million PBMNCs were obtained from the

isolation process, then subsequently suspended in endothelial cell basal media-2 (EBM-

2) (Lonza, Walkersville, MD) supplemented with 20% fetal bovine serum, human

fibroblast growth factor (hFGF), vascular endothelial growth factor (VEGF), human

epidermal growth factor (hEGF), human recombinant insulin-like growth factor (R3-IGF-

1), ascorbic acid, gentamicin, amphotericin, and heparin. Henceforth, this fully

supplemented EBM-2 is referred to as endothelial cell growth media-2 (EGM-2).

Page 150: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

150

Culture and Differentiation of Endothelial Progenitor Cells

The PBMNCs were plated at a density of 2-3X105 cells/cm2 onto fibronectin-

coated tissue culture polystyrene plates (BD Biosciences, Franklin Lakes, NJ) in

complete cell culture media as described above. The culture was maintained for 2-6

weeks and the cells were visually assessed to observe colony formation every day

using an inverted phase-contrast microscope (Nikon TE2000U, Melville, NY). The first

appearance of tightly packed, cobblestone morphology, human endothelial-like (HE-like)

cells in a given field of view was recorded as indicative of EC differentiation. The

differentiated cells were sub cultured and utilized within passages 1-3. Human umbilical

vein endothelial cells (HUVECs) (Lonza) were used as a positive control for mature EC

functionality. HUVECs were cultured in EGM-2 (with only 2% FBS) and utilized within

passages 3-5.

Immunocytochemical Assessment of Differentiated Endothelial Cells

The endothelial cell phenotype of the differentiated cells was confirmed using

immunocytochemical analysis (herein after referred to as human endothelial-like cells

(HE-like cells)). Briefly, HE-like cells were fixed in 4% paraformaldehyde, and then

blocked using 3% bovine serum albumin for 1h. The cells were incubated overnight at

4⁰C with a primary antibody specific to von Willebrand factor (vWF) (diluted 1:100)

(Abcam, Cambridge, UK) and CD31 (diluted 1:100) (Abcam). The cells were

subsequently incubated with a fluorescein isothiocyanate tagged isotype-matched

secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA) and the nuclei were

counter-stained using Hoechst 33342 (Thermo Scientific). Fluorescent images were

obtained using an inverted fluorescent microscope (Nikon TE2000U) (Nikon, Melville,

NY).

Page 151: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

151

Simulated Microgravity Culture of HE-Like Cells

Cytodex 1 beads (Sigma Aldrich, St. Louis, MO) were swollen and sterilized

using manufacturer’s instructions.127,282,288 The beads were coated with fibronectin

(25ng/ml) (Sigma Aldrich) to promote cell adhesion. HE-like cells obtained using the

above-mentioned procedure, were seeded on the beads at a density of 3 X 106 cells per

250 mg of beads in ultra-low attachment cell culture plates. The culture was maintained

for up to four days until complete confluence of cells on beads was obtained. The cell

coated beads were then transferred into the bioreactor chambers (Synthecon, Houston,

TX) as shown in Figure 7-1 A&B and cultured under simulated microgravity conditions.

Figure 7-1. Bioreactor set-up and operation. (A) Synthecon RWV bioreactor set-up (B) RWV bioreactor detachable chamber (C) Representative 20X phase contrast image of HE-like cells cultured on cytodex-1 microcarrier beads. Scale bar is 100 µm. In panel C, the arrows indicate cell covered beads.

Briefly, the bioreactor was set to rotate at an initial speed of 8.5 rpm, to suspend the

cells under simulated microgravity with minimum shear stress in a cell culture incubator

with 5% carbon dioxide (CO2). For all experiments, as a static control, the cell coated

beads were placed into the bioreactor chambers in a cell culture incubator, however, the

chambers were not rotating and thus were maintained under normal gravity.

Subsequently, the cultures were maintained for up to 6 days. About half of the cell

culture media in the chamber was replaced every other day.

Page 152: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

152

Cell Proliferation Assessment

Cell proliferation was assessed via changes in DNA concentration of HE-like

cells cultured both under simulated microgravity and normal gravity over time. The

dsDNA was quantified using Quant-it picogreen dsDNA assay kit (Thermo Scientific)

following manufacturer instructions. Briefly, HE-like cells on beads, exposed to

simulated microgravity were collected at 1, 2, 4, and 6 days of culture from the

bioreactor chamber. Correspondingly, samples were collected from normal gravity

controls at the same respective time points. Then, the cells were lysed using 0.1 %

triton-X to collect and quantify the intracellular DNA. The DNA concentrations of lysates

were quantified and plotted versus time.

Gene Expression

To determine the effect of culture conditions on gene expression of hsp70 by the

HE-like cells, quantitative reverse transcriptase polymerase chain reaction (qRT-PCR)

was conducted. After 2 days and 4 days of culture, total RNA was isolated from the HE-

like cells cultured under normal and reduced gravity conditions, using the RNeasy Mini

Kit (Qiagen, Valencia, CA) according to manufacturer instructions. Subsequently iScript

cDNA Synthesis Kit (Bio Rad, Hercules, CA) was used to generate first-strand cDNA.

The RNA sequences of human glyceraldehyde 3-phosphate dehydrogenase (GAPDH)

and heat shock protein 70 kDa A 4 (HSPA4) were identified and selected from the

National Center for Biotechnology Information (NCBI) gene database, and

corresponding primers for each of the genes were obtained from Integrated DNA

Technologies ( IDT, Coralville, IA). qRT-PCR was run on the Bio Rad CFX Connect (Bio

Rad, Hercules, CA) using Maxima SYBR Green (Thermo Scientific, Waltham, MA) to

quantify the expression of each target gene. Melt curve analysis was run to ensure

Page 153: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

153

amplification of non-specific PCR products did not affect the quantification results.

Results for gene expression were normalized using GAPDH. The data obtained was

derived from cells from a single donor population.

Endothelial Cell Secretory Function

The amount of nitric oxide (NO), interleukin-6 (IL-6), and tissue plasminogen

activator (tPA) secreted into the culture medium by the HE-like cells was assessed after

2 and 4 days of culture in both normal and simulated microgravity. Quantification was

obtained utilizing the total NO assay kit (Cayman Chemical, Ann Arbor, MI), Human IL-

6, and Human t-Plasminogen Activator/tPA Quantikine ELISA Kits (R and D systems,

Minneapolis, MN) respectively, following manufacturer protocols. Briefly, the

concentration of NO released into the medium was quantified based on the enzymatic

conversion of nitrates to nitrites and the photometric detection of total nitrite

concentration via the Greiss reaction. The concentration of NO was normalized to cell

density using the dsDNA concentration of the cells and expressed in terms of nitrite

concentration (µM) per pg of dsDNA, to draw comparisons between times in simulated

microgravity.

Plasma Clotting Kinetics

The plasma clotting kinetics of the HE-like cells was assessed using a modified

protocol previously reported230,270. Briefly, human allogenic platelet poor plasma was

obtained via centrifugation from whole blood. HE-like cells on beads cultured for 2 and 4

days, both under simulated microgravity as well as under normal gravity were exposed

to the 250 µl of the plasma recalcified by the addition of 0.025 M calcium chloride

(CaCl2). Following which, the absorbance at 405 nm was recorded using a Synergy H1

microplate reader (Biotek, Winooski, VT). As the plasma clots it becomes more opaque,

Page 154: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

154

thus an increase in absorbance with time is indicative of clot formation. HUVEC coated

microcarrier beads served as a positive control for anti-thrombogenic function of mature

ECs. The full kinetic profile of clot formation as well as the time taken to reach maximum

absorbance, indicative of full clotting, was recorded and reported for each

sample/control.

Statistical Analysis

All data are reported as mean +/- standard deviations. A student 2-sample t-test

was employed to compare two numerical means. Analysis of variance (ANOVA) with

Tukey’s multiple comparison post hoc analysis was used to analyze and identify

significant differences between 3 or more means. P-values <0.05 are reported as

significant.

Results and Discussion

The physiological threats faced by astronauts as a direct consequence of human

space flight have been identified and studied extensively in the recent past135,139,265. The

cardiovascular system of astronauts has been known to be affected by orbital space

flight with the changes manifesting as cardiovascular deconditioning135,266,267. From a

clinical perspective, deconditioning is observed as a: (1) decrease of circulating blood

and interstitial fluid volumes, (2) decrease of arterial blood diastolic pressure, (3)

decrease of ventricular stroke volume, (4) decrease of estimated left ventricular mass

and (5) a resetting of the carotid baroreceptors. The deconditioning effects of

microgravity manifest itself mostly upon the reentry to Earth268. Maintenance of vascular

homeostasis lies in the proper function of the vascular cells, specifically, the ECs that

line the vessel wall. This work is focused on understanding the effects of short term

simulated microgravity on the downstream functions of ECs derived from whole blood.

Page 155: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

155

Previous work has shown the exposure of mature ECs to simulated microgravity for as

little as a few minutes to a few days results in structural and morphological changes as

well as modifications to both gene and protein expression 143,263,285,288–290,300. These

results have been observed and validated in both space flight experiments as well as

ground based microgravity simulators as was used here. However, there is very limited

literature on the effect of altered gravity conditions on blood-derived circulating

endothelial progenitor cell (EPC) derived ECs. EPCs are a small population of

progenitor cells that reside in bone marrow and, upon stimulation, are released into

circulating peripheral blood. EPCs along with the endothelium are critical to the

maintenance of vascular homeostasis, which includes regulating the hemodynamics of

blood flow, vessel wall tone, and the secretion of important signaling molecules270,274,279.

Previous studies have shown the ability of circulating progenitor derived ECs to function

in a similar manner to mature ECs including their ability to regulate clotting mechanisms

in vitro85,230,243,270,299.85,230,243,270,299. Despite the conflicting reports on the suitability of

using EPCs for the study of cardiovascular diseases276, this data shows the promising

potential of EPCs to act as a repair cell population in cardiovascular regeneration.

Therefore, this work is an important stepping stone towards understanding the role

played by EPCs for the onset of cardiovascular deconditioning during space travel and

to develop possible cell based therapies. As a foundation for the work, the group has

previously shown the effect of simulated microgravity on the differentiation and

downstream anti-thrombogenic functions of porcine endothelial progenitor cells230.

Page 156: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

156

In this report, circulating progenitor cells contained within the PBMNC fraction of

whole blood were isolated then cultured in the presence of vascular endothelial growth

factor (VEGF) to induce differentiation along the endothelial lineage84,85,230,270.

Differentiation into endothelial phenotype was confirmed by the formation of colonies

with EC specific ‘cobble-stone’ morphology (HE-like cells) as shown in Figure 7-2 A.

Figure 7-2. Isolation of PBMNCs and differentiation into ECs: (A) Representative phase contrast image of PBMNC derived colony formation of HE-like cells, Scale bar 200 µm. (B) Representative phase contrast image of characteristic endothelial ‘cobble-stone’ morphology of HE-like cells in subsequent culture (20X). Representative 20X fluorescent images of positive immunocytochemical staining of HE-like cells for (C) CD31 and (D) von Willebrand factor, both shown in green, nuclei were counterstained with Hoechst 33342 shown in blue. Panel B-D Scale bar is 50 µm.

Colony formation was observed between 2-6 weeks of culture. In all cases the

observation for colonies was concluded after 50 days. The process was repeated for

three populations; each from a different donor. The data reported are representative of

Page 157: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

157

these three different populations. The endothelial phenotype of these cells were

confirmed and validated by the maintenance of the characteristic ‘cobble-stone’

morphology in subsequent culture as shown in Figure 7-2 B and positive

immunocytochemical staining for CD31 as shown in Figure 7-2 C, and vWF, as shown

in Figure 7-2 D both of which are specific for EC phenotype.

To simulate microgravity culture conditions, HE-like cells were seeded onto

fibronectin coated microcarrier beads. A RWV bioreactor system, designed originally by

NASA was used to simulate a reduction in time-averaged gravitational vector acting on

these cells, reducing it to about 10-2 g. For simplicity, this reduced gravity condition is

referred to as microgravity133,136,263. Although RWV systems do not mimic actual

microgravity as experienced by astronauts, however, it is a useful tool to provide insight

into the behavior of various cell systems in space127,282.The culture was maintained by

adjusting the rotational speed just enough to maintain the vessel wall and the medium

containing cells bound to microcarrier beads rotating at the same speed.

One of the primary and most important functions of the HE-like cells is their

ability to adapt to changing conditions and to continually proliferate. The effect of

simulated microgravity (µg) on differentiated HE-like cells and compared to the kinetics

of proliferation under normal gravity (1g) conditions. The proliferative capacity of the

HE-like cells was assessed by quantifying the change in DNA over 1, 2, 4 and 6 days of

culture. The results indicated the concentration of DNA in the normal gravity (1g)

cultured cells initially was higher than the simulated microgravity (µg) up to 2 days of

culture (p<0.05) as shown in, Figure 7-3. [dsDNA concentration (µg/ml) after 1 day: 1g

0.098 +/- 0.002, µg 0.073+/- 0.013 p<0.05; after 2 days: 1g 0.110+/-0.005, µg 0.093+/-

Page 158: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

158

0.007 p<0.05]. However, the cells cultured under simulated microgravity caught up by

day 4 and showed no significant difference in DNA content. [dsDNA concentration

(µg/ml) after 4 days: 1g 0.116 +/- 0.005; µg 0.113 +/- 0.003 not significant]. After 4 days,

the HE-like cells under simulated microgravity increased there rate of proliferation as

indicated by a higher concentration of DNA when compared to the normal gravity

control at day 6. [dsDNA concentration (µg/ml) after 6 days: 1g 0.151+/- 0.010, µg

0.173+/- 0.003 p<0.05].

Figure 7-3. Proliferation kinetics: Representative proliferation kinetics curve HE-like cells cultured under microgravity (µg) vs cells maintained in normal gravity (1g) as measured by DNA concentration in cell lysates. Data shown are Mean +/- SD; n=3.

The results showed an initial decrease in cell growth which was reversed over

increased time in culture. A similar trend in proliferation has been shown for a mature

EC population upon exposure to simulated microgravity285,301. This data indicates that

HE-like cells function similar to mature EC populations when exposed to simulated

microgravity, thus enhancing their ability to proliferate. Additionally, this trend also

indicates a potential recovery effect in the proliferative capacity of these ECs and HE-

like cells. Such a proliferation effect has been reported in PBMNCs 291 and other mature

Page 159: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

159

EC populations136,263,285,302 .This trend also indicates the potential ability of these ECs to

sense the change in gravitational conditions as stress and their ability to recover from

this stress by enhancing proliferation and cell growth302.

Further the ability HE-like cells to sense simulated microgravity as a stressful

condition and adapt to it by regulating the levels of genes encoding stress proteins such

as hsp70 was investigated. The regulation of gene expression of HSPA4 via RT PCR.

The HSPA4 gene belongs to the family of heat shock protein 70 was quantified. Heat

shock proteins (hsp) have been known to play an important role in cell protection and

repair upon stress as well as under certain non-stressful conditions302–304. More

specifically, hsp70 has been identified as a molecular chaperone which can inhibit

apoptosis. Mechanistically hsp70 inhibits apoptosis in cells by preventing the

constitution of the apoptosome and by protecting against the apoptogenic effects of

apoptosis inducing factors (AIF) targeted to the extramitochondrial compartment304,305.

The results showed that simulated microgravity-cultured cells up-regulated gene

expression of HSPA4 which belongs to the family of genes encoding hsp70 protein,

after 2 days of culture as shown in Figure 7-4. [After 2 days: 1g 0.496+/- 0.129, µg

1.0+/- 0.306, p<0.05]. However, after 4 days in culture, there was no statistical

difference in the level of HSPA4 between simulated microgravity and normal gravity.

[After 4 days: 1g 0.346 +/- 0.154; µg 0.726+/- 0.101, not significant]. The data generated

was from a representative population of HE-like cells isolated from a single donor.

Previous studies have shown the ability of mature EC populations to sense simulated

microgravity as a stressful condition and upregulate proteins such as hsp70 and

maintain their ability to proliferate133,263,301,302.

Page 160: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

160

Figure 7-4. Gene expression. Representative relative gene expression of HSPA4 by microgravity (µg) cultured HE-like cells vs cells maintained in normal gravity (1g) over 2 and 4 days of culture. Data shown are Mean +/- SD; n=3. N.S. indicates no statistically significant difference (p > 0.05) and * indicates a statistically significant increase (p<0.05).

Consistent with this trend, the results indicated that culture under simulated

microgravity conditions led to an initial upregulation of gene expression of HSPA4 over

2 days of culture but by day 4 the levels were similar between the normal and simulated

microgravity conditions. This data provides mechanistic insight into the ability of the HE-

like cells to survive and proliferate under “stressful” conditions such as simulated

microgravity by regulating stress proteins such as hsp70.

Another important molecule that regulates EC function is nitric oxide (NO). NO is

an important regulator of various important functions of the endothelium such as: cell

proliferation, angiogenesis, differentiation, and anti-thrombogenicity 299,306.Results

indicate that HE-like cells cultured under simulated microgravity for 2 days released

higher amounts of soluble NO when compared to the respective normal gravity controls.

[NO concentration (µM/pg of dsDNA): 1g 0.64+/- 0.308, µg 1.77+/- 0.202 p<0.05].

However, after 4 days of culture, the level of NO release was statistically similar

Page 161: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

161

between both groups. [NO concentration (µM/pg of dsDNA): 1g 2.49 +/- 0.050; µg 2.51

+/- 0.079 not significant] as shown in Figure 7-5 A.

Figure 7-5. Secretory functions of cells. Representative results of quantification of release of (A) Nitric oxide and (B) Interleukin-6 by microgravity (µg) cultured HE-like cells vs cells maintained in normal gravity (1g) over 2 and 4 days of culture. (Data shown are Mean +/- SD; n=3). N.S. indicates no statistically significant difference (p > 0.05) and * indicates a statistically significant increase (p<0.05).

Previous studies have reported a simulated microgravity-mediated increase in

endothelial nitric oxide synthase protein levels as well increased NO production at 48 h

and 72 h of simulated microgravity exposure respectively by mature ECs300. Also, a

study by Spisni et al. (2006) 307 reported no change in endothelial nitric oxide synthase

(eNOS) or inducible nitric oxide synthase (iNOS) protein levels yet an increase in NO

production at 24 h of simulated microgravity exposure. This increased level of NO may

also indicate another possible recovery mechanism of the cells to adapt to the changing

gravitational forces 285,301,302,308.Also more importantly, an increase in expression of NO,

a known vasodilator, leads to increased relaxation and lowering of blood pressure which

is known to disrupt sympathetic flow, leading to orthostatic intolerance306,308.Hence, this

may also provide a possible insight into cardiac dysfunction caused by space travel.

The relationship between endothelial dysfunction and inflammatory response

leading to cardiovascular complications such as atherosclerosis has been widely

Page 162: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

162

studied in the recent past246,299,309. Immune dysfunction in astronauts in space

environment has also been studied extensively288,310,311. A potential anti-inflammatory

response to short term culture under simulated microgravity by the down regulation of

pro-inflammatory cytokines has been reported by multiple studies133,300. These findings

have also been implicated to be mediated by changes in cytoskeletal arrangement in

simulated microgravity300. Assessment of the release of soluble IL-6, a major pro-

inflammatory cytokine, show that culture under simulated microgravity conditions for 2

and 4 days led to significant decrease in secretion of soluble IL-6 in the cell culture

medium when compared to normal gravity cultured controls. [IL-6 concentration (pg/ml)

after 2 days: 1g 46.3+/-1.0, µg 11.71+/-0.88 p<0.05; after 4 days: 1g 97.013 +/- 0.592;

µg 53.697 +/- 9.196 p<0.05] as shown in Figure 7-5 B.

Consistent with the literature reports for mature ECs, the results showed a

decrease in expression of IL-6, an important pro-inflammatory and anti-proliferative

cytokine 312. It is noteworthy that the inverse relationship between NO and IL-6 that was

seen in this study has previously been reported for ECs313,314. Hence the

downregulation of IL-6 may play an important role in the response of the HE-like cells to

simulated microgravity, since low amounts of IL-6 might contribute to cell growth as well

as to the enhancement of NO synthesis. Similarly the ability of hsp70 to inhibit the

synthesis of pro-inflammatory cytokines in ECs has also been reported315–317. This

altered immune function observed for the HE-like cells not only has implications in their

regulatory role regarding the overall immune response of the body during space travel,

but also additional implications on the effect of space travel on overall cardiovascular

health.

Page 163: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

163

The anti-thrombogenic behavior of ECs is important to maintain a healthy

vascular system and to prevent cardiovascular complications such as thrombosis269.

Healthy endothelium secretes various factors that are important in regulating

coagulation and fibrinolysis 243,299.Tissue plasminogen activator (tPA), an important

protein released by ECs, has been shown to be a main factor in the activation of the

fibrinolytic system, which is involved in dissolution of clots. Specifically, tPA activates

the conversion of clot bound plasminogen to plasmin which leads to clot lysis318,319. An

increase in expression of tPA has been directly correlated with increased bleeding while

a low expression of tPA has been implicated in thrombosis320. Quantification of the

release of soluble tissue plasminogen activator (tPA) into the surrounding cell culture

medium showed that tPA released by HE-like cells cultured under simulated

microgravity conditions for 2 and 4 days were significantly lower than their respective

normal gravity cultured controls [tPA concentration (ng/ml) after 2 days, 1g 4.572+/-

0.370, µg 2.01+/- 0.263 p<0.05; after 4 days: 1g 13.26 +/- 0.557; µg 9.867 +/- 0.528

p<0.05] as shown in Figure 7-5 C.

The data assessing the expression of tPA released by HE-like cells indicates

simulated microgravity mediated an increase in expression of tPA over 2 and 4 days of

culture. This indicates that exposure of HE-like cells to simulated microgravity can

potentially decrease the anti-thrombogenic potential of the cells. In order to more

directly study the anticoagulative functions of the HE-like cells, the ability of these cells

to delay clotting of human blood derived allogenic plasma was assessed using a

modified version of plasma clotting assay was performed. This assay provided a robust

and direct method to assess and compare the anti-thrombogenic potential of HE-like

Page 164: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

164

cells cultured under different gravity conditions. Once clotting was activated, progress to

full clot formation was assessed spectrophotometrically by continuous monitoring of the

change in absorbance over 30 min.

Figure 7-6. Plasma clotting kinetics. Recalcified plasma clotting kinetics for human plasma incubated with HE-like cells cultured in normal vs. simulated microgravity for (A) 2 days; and (B) 4 days. Data show absorbance plotted versus time. Arrows indicate the rightward shift in the normal gravity conditions, indicative of delayed clotting. SD error bars have been omitted for visual clarity. (Data shown are Mean +/- SD; n=3). (C) Representative result showing quantification of the time taken for recalcified plasma to clot (in minutes) when incubated with HE-like cells cultured for 2 and 4 day in normal vs simulated microgravity. Data shown are Mean +/- SD; n=6. N.S. indicates no statistically significant difference (p > 0.05) and * indicates a statistically significant increase (p<0.05).

The clotting profiles of recalcified plasma incubated with HE-like cells cultured in

both simulated microgravity as well as normal gravity for both 2 and 4 days is shown in

Figure 7-6. HE-like cells cultured in normal gravity exhibited a significant delay in the

onset of clotting as indicated by the rightward shift in the kinetic profile, indicating a

degree of anti-thrombogenic function. This contrasted with the kinetic profile of plasma

clotting when incubated with HE-like cells cultured under simulated microgravity, which

show an earlier onset to clot formation. These trends are seen at both 2 days and 4

days of culture as shown in Figure 7-6 A and B. As a control, CaCl2 was omitted from a

Page 165: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

165

subset of samples, and the plasma did not clot (data not shown). We also quantified

and reported the time taken to reach maximum absorbance indicating complete clotting

of plasma on the cell covered beads. The results indicate that recalcified plasma clots

faster when incubated with HE-like cells cultured under simulated microgravity as

compared to the longer time it takes plasma to clot when incubated with HE-like cells

cultured under normal gravity as shown in Figure 7-6 C [clotting time (min): with 2 day

cultured HE-like cells: 1g 14.31+/- 2.199, µg 10.001+/- 1.395 p<0.05; with 4 day

cultured HE-like cells: 1g 16.569 +/- 1.357; µg 12.456 +/- 1.457 p<0.05]. HUVEC coated

beads were utilized as the control population of mature ECs, and showed the most

delayed plasma clotting (19.3 +/- 1.9 min) (Data not shown). Similarly, naked beads

devoid of HE-like cells were also assessed, and they showed no delay in plasma clot

formation (data not shown). The clotting data corroborated the assessment of tPA

secretion, in that HE-like cells exposed to simulated microgravity were not able to delay

clot formation as well as the normal gravity cultured controls over 2 and 4 days of

culture. Also, both populations, normal and simulated microgravity show anti-

thrombogenic function, however, with exposure to simulated microgravity, this function

was hindered. This data collectively shows that exposure to simulated microgravity

negatively influences the anti-thrombogenic function of HE-like cells relative to those

cultured under normal gravity conditions. This inhibition of anti-thrombogenic

functionality of the cells potentially provides insight into thrombosis and other

coagulation processes in the body which have been known to directly cause

cardiovascular deconditioning. Although the ability of simulated microgravity to directly

affect platelet functions has been shown before,144 to the knowledge, the effect of

Page 166: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

166

simulated microgravity on direct anti-thrombogenic properties of EPC derived ECs, such

as inhibition of plasma clotting has not been reported thus far.

It is important to note that decreased production of inflammatory cytokines such

as IL-6 as well as increased production of NO have been known to synergistically inhibit

clotting processes in the body85,299,306,309. However, in this study, it was observed that

although there was a significant reduction in the release of IL-6 and an increase in

release of NO under simulated microgravity, the cells were able to induce faster clotting

of plasma. It is also interesting to note that although culture under simulated

microgravity conditions affects various secretory and proliferative mechanisms of the

HE-like cells, these functions seem to not influence the downstream ability of the cells to

delay clotting. This indicates the need to study extensively the effect of simulated

microgravity on various alternate and synergistic mechanisms involved in both

inflammatory as well as thrombogenic processes. Also, the relatively small sample size

(n=3, donor EPC populations) limits the detection of small statistical differences

between samples. Nevertheless, this report provides a useful insight into the modulation

of clotting mechanisms as well as secretory and proliferative ability of blood derived

ECs when exposed to reduced gravity conditions. Since these cells are in direct contact

with circulating blood, this study will prove to be particularly important when assessing

their role in cardiovascular deconditioning during space travel, as well as to develop

countermeasures to ameliorate or possibly reverse this effect.

Summary

The disruption of normal functioning of the endothelium has been shown to

contribute significantly in weightlessness-induced cardiovascular deconditioning during

space flight267. The focus of this study was to bridge the gap in knowledge on the effect

Page 167: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

167

of altered conditions such as microgravity on circulating stem cells as they differentiate

into endothelial cells. Circulating stem cells have been known to contribute significantly

to vascular repair mechanisms in the body277,279. The motivation for this work was to

study and understand the behavior of these cells in a low shear stress environment

such as simulated microgravity to assess their therapeutic potential for vascular repair

and regeneration.

In this study a NASA designed, rotating wall vessel (RWV) bioreactor that mimics

certain functional aspects of microgravity on earth (Synthecon, Houston, TX) was

utilized. The RWV was designed and developed at NASA’s Johnson Space Center to

simulate the effects of microgravity on cells in a ground-based culture system134,282. The

results obtained using simulated microgravity conditions utilizing the RWV have been

confirmed and validated by experimental models in space flights to outer space133,302.

The first part of the above described work involved the effect of simulated

microgravity on the differentiation of EPCs pre-exposed to simulated microgravity. This

report also studied the downstream function of the differentiated cells in terms of their

migration and proliferative capabilities, release of soluble factors such as PGE2 and

NO, and ability to regulate clotting mechanisms in the body. While both the migration

and proliferation of cells were inhibited by microgravity exposure, prolonged exposure of

EPCs to microgravity resulted in decrease in the migration abilities of EPC derived EC

and the proliferative capacity of the ECs seemed to recover on longer time of exposure.

The initial decrease and subsequent increase in release of soluble factors such as NO

and PGE2 reaffirmed the possibility of recovery mechanisms being involved on

prolonged exposure to weightlessness. Finally the assessment of whole blood and

Page 168: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

168

plasma clotting, indicated that microgravity pre exposure at the progenitor stage caused

inhibition, in the ability of PE-like cells to delay blood or plasma clotting while this effect

was again nulled with increase in time of exposure of EPCs to microgravity. This data

collectively provides some insight into the role of short term exposure to low shear

stress environment due to simulated microgravity, on cardiovascular deconditioning

during space travel. Especially, the effect of microgravity on anti-thrombogenic

properties of the endothelium has not been reported before. Encouraged by the results,

the study was further extended to include human blood progenitor derived endothelial

cells. The goal was to investigate and draw comparisons between the effect of

simulated microgravity on vascular regeneration mechanism of EPCs and ECs when

exposed prior and post differentiation respectively.

This was demonstrated in Chapter 6. The results from the study show that the

populations of human blood EPC derived ECs when exposed to simulated microgravity

proliferate faster, release higher amounts of soluble NO and lower amounts of pro-

inflammatory cytokines such as IL-6, when compared to their normal gravity cultured

controls. This can be possibly attributed to the ability of the cells to sense simulated

microgravity as a “stressful” condition and adapt by upregulating gene expression of

hsp70. However, these secretory as well as proliferative functions had no effect on the

anti-thrombogenic functions of these cells. Their anti-coagulative and anti-thrombogenic

ability as assessed by both upregulation of tPA as well their ability to delay plasma

clotting, were impaired upon exposure to simulated microgravity.

It is important to note here that even though simulated microgravity is considered

a low shear environment, further investigation have shown that a small amount of shear

Page 169: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

169

stress is experienced by particles in rotation in simulated microgravity bioreactors282,321.

This shear stress varies with particle size and the velocity of rotation. The average

shear stress on a microcarrier bead as used in the study based on mathematical

calculations and estimations has been shown to be < 0.5 dynes/cm2 as described in

detail in the background section. The shear stress is estimated to be even lower on cells

without a carrier being rotated in the field. Although this level of shear is tremendously

lower that arterial shear stress or even venous shear stress values, its effect still needs

to be considered. The endothelium is very sensitive to variations in the characteristics of

flow that in turn cause changes in shear stresses; which manifest as pathologic

changes in the artery wall.

While much work has been done to study the response of ECs as well as EPCs

under physiological and higher shear stress conditions, effects of low shear stress ( <1

dynes/ cm2) has not been well characterized. Recent work by Mazzolai et al. (2011),

characterized human late outgrowth EPCs under low shear stress ( ~ 0.3 dynes/ cm2)

and physiological shear stress values (~ 6 dynes/ cm2)322 . Their results showed that

under low shear the expression of tissue factor mRNA was significantly upregulated.

While the expression of tissue-type plasminogen activator (tPA), urokinase (u-PA) and

monocyte chemotactic protein (MCP1) were downregulated. These results implicated a

possible shift of these cells towards a pro-coagulant cell type. This shift was also shown

to be dependent on the direction of shear. These results are comparable to the current

study which showed reduced anti-thrombogenic potential. However, Mazzolai et al. also

noted no significant effect on inflammatory response via regulation of IL-6 which was in

contrary to what was observed in simulated microgravity culture where inflammatory

Page 170: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

170

cytokine IL-6 was downregulated significantly. This trend has however been seen with

mature ECs in simulated as well as real microgravity previously. Moreover, the current

study also shows consistent initial delay and then recovery of proliferative potential of

both EPCs and EPC derived ECs which is not observed with application of shear stress

alone. This could possibly be the differences in shear experienced by cells under

laminar flow in comparison with cells under rotation in a RWV bioreactor. Since

simulated microgravity produces the unique effect of mimicking weightlessness, the

study of cellular behavior under this condition is not directly comparable to shear stress

induced cell response.

Overall this study serves as the first step towards understanding the ability of

these repair cells to regulate clotting mechanisms and cardiovascular health in

response to macroscale cues provided to them by altered growth conditions. Further

studies to assess alternate and synergistic mechanisms involved in regulation of

cardiovascular health as well as inflammatory dysfunction under microgravity are

required to develop possible cell based countermeasures to prevent cardiovascular

deconditioning due to space travel. This approach also has the potential to be applied to

develop strategies for vascular regeneration on earth using EPCs.

Page 171: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

171

CHAPTER 8 SUMMARY, CONCLUSIONS AND FUTURE WORK

Summary of Results

Cardiovascular diseases are the leading cause of death worldwide, with more

than 60% occurring within United States itself1. Momentous improvements in clinical

care for patients suffering from complications due to cardiovascular diseases have been

made through the years. Significant efforts have been focused on perfecting

pharmaceutical and transplant based therapies to treat these diseases. However, the

chronic scarcity of donor organs fit for transplantation has led to increased efforts

towards developing novel tissue engineering based strategies to develop functional

replacements for diseased or damaged parts of the vasculature6,26,323.

Tissue engineering strategies are based on the premise that the functional cell is

the true ‘tissue engineer’17. Therefore, these strategies make use of ways to modulate

cellular response by providing appropriate cues to the cells. Given the complexity of the

vascular system itself, the cues presented must be multi-dimensional and at different

length scales of interaction. While many such cues have been studied historically, there

are many unknown stimuli whose effect is yet to be explored and exploited. With this

motivation in mind, the goal of this dissertation was to study the underlying mechanism

of specific cell-material interactions at different scales (nano, meso and macroscales),

to develop novel strategies to repair, preserve and regenerate vascular tissue.

Starting at the nanoscale, the first part of this work was focused on the use of a

supramolecular aptamer assembly as a receptor agonist. This subcellular interaction

was characterized by studying the activation of VEGFR2 receptor in endothelial cells by

the aptamer assembly. Activation of the VEGFR2 receptor has been shown to facilitate

Page 172: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

172

various processes such as proliferation, migration, neoangiogenesis and differentiation

in endothelial cells and various stem cell populations39,324. These positive outcomes of

the activation event have been exploited in designing pro-angiogenic therapies via

targeted activation of VEGFR2 for vascular tissue engineering to cause repair and

regeneration of vascular tissue37,325.

The results presented in this study show the design and evaluation of a divalent

aptamer assembly consisting of two monomeric units tethered together using a PEG

chain. This novel strategy of using nucleic aptamers based divalent assembly to target

VEGFR2 was effective in triggering the activation cascade in these receptors in

endothelial cells. The assembly was also able to cause positive downstream angiogenic

response which proves that this is a promising strategy to modulate cell behavior

favorably for vascular tissue engineering and regenerative medicine, where controlling

cell processes is of paramount importance. The ability to modulate a functional

response such as cell growth, differentiation and angiogenesis is critical for success in

these areas. Therefore, this novel, specific and targeted strategy has tremendous

potential towards designing novel nanoscale biomaterial cues for vascular tissue

engineering.

Moving up on the scale of interactions, the next part of this work consisted of

studying cues designed at the meso scale. The biological mesoscale range has been

described to be inclusive of biological structures ranging from 10 to several 100 microns

in size14,26. This includes interaction of cells with surrounding biomaterial scaffolds that

house the cells. Biomaterial environments provide a variety of physical, mechanical,

chemical as well as structural cues that can be altered for cardiovascular applications15.

Page 173: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

173

This section covered two classes of cardiovascular biomaterial applications, namely,

small diameter vascular grafts and vascular stents. This work was focused on studying

the interaction of these biomaterials with in vitro models of the vascular environment to

understand and develop strategies to repair damaged vasculature and enable

regeneration.

First part of the study, was focused on development of a novel bilayered,

construct for application as a vascular graft. The graft was designed to mimic the

biological and architectural aspects of native blood vessels. The design consisted of two

layers, a solid luminal layer made of elastomeric, biodegradable POC and a fibrous

medial layer consisting of a mixture of Collagen, POC and elastin. The rationale behind

the design is to provide a solid lumen for the formation of a monolayer of endothelial

cells and fibrous medial layer enriched with ECM proteins to enable formation of an

interconnected network of SMCs. The individual layers of the graft were separately

fabricated and assessed for cell attachment and proliferation. The constructs

mechanical properties were also assessed. Overall the results showed that the graft

was biocompatible and was able to support attachment and growth of vascular cells.

More interestingly, the mechanical properties evaluation revealed that the compliance of

the fabricated graft matched native artery values. This property is very important to

prevent implant failure due to mismatch in mechanical properties. This graft is currently

being developed to recruit circulating stem cells to promote integration into vasculature.

The second part of the study evaluated the hemocompatibility of an Mg-Ca-Sr

alloy to determine its utility as a candidate material for vascular applications. Overall,

this study showed that the alloy is biocompatible and invokes very low cytotoxic

Page 174: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

174

response when cultured with a mature vascular endothelial cell population. The alloy

sample was hemocompatible and exhibited excellent thromboresistance as indicated by

low hemolytic percentage as well as delayed and reduced clotting when in contact with

whole blood. The alloy also did not cause significant platelet adhesion and activation.

Finally, it also does not induce significant release of pro-inflammatory factors confirming

minimal inflammatory response. This data collectively confirms the suitability of the Mg-

Ca-Sr alloy for potential application as a vascular stent material.

The studies combined investigated the effect of biomaterial structural cues on

the meso scale on their ability to be used in vascular environments. The studies

involved evaluating some important characteristics of these materials in terms of their

biocompatibility, hemocompatibility, ability to support vascular cell types, mechanical

and biochemical behavior. These factors are essential in preventing vascular damage

due to complications such as thrombosis, toxicity as well as, fostering healthy

remodeling and regeneration of vasculature. Combined, these strategies widen the

knowledge in biomaterial-cell interaction and are preliminary steps towards developing

suitable biomaterials for vascular tissue engineering.

The final part of this dissertation was focused on macroscale systemic cues that

provide a unique environment for modulating response in vascular cells. The dynamic

nature of the cardiovascular system and the role of its constituent cell systems has been

studied extensively. The endothelium which lines the inner layer of blood vessels is

especially critical to its normal functioning269. It is this layer that actively and dynamically

controls repair and regeneration processes occurring within the vascular network. This

layer has been shown to respond to physiological mechanical cues, such as shear

Page 175: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

175

stress, cyclic stretch, and alterations to gravity which cause them to undergo significant

morphological and functional changes260,264.

One such specialized macro environmental model is microgravity in space flight,

which provides a unique opportunity to study the effect of gravitational changes on cells.

The negative effects of space travel on cardiovascular health has been well

documented in the past. A common feature of the effects of altered gravitational

conditions on different cellular systems is that the affected tissues rely on tissue-specific

stem cells for regeneration and repair. Understanding and replicating the behavior in

such a condition can lead to development of stem cell based therapy for cardiovascular

degeneration on earth. A recent study by Chiu et al., has shown that human cord blood

stem cells when cultured under simulated microgravity proliferated and trans-

differentiated into 3D structures resembling vascular tubules while exhibiting vascular

endothelial phenotype expressions140. Many such studies on the effect of stem cells

under alternate culture environments especially simulated microgravity is currently

being studied to develop regenerative strategies. Current research is also studying the

hypothesis that cardiovascular deconditioning due to space travel may potentially trigger

faster maturation of stem cells and therefore enhancing their regenerative potential. The

focus of this study was to bridge the gap in knowledge on the effect of altered conditions

such as microgravity on circulating stem cells as they differentiate into endothelial cells.

Circulating stem cells have been known to contribute significantly to vascular repair

mechanisms in the body270,326. The motivation for this work was to study and

understand the behavior of these cells in a low shear stress environment such as

Page 176: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

176

simulated microgravity to assess their therapeutic potential for vascular repair and

regeneration.

The first part of the above described work involved the effect of simulated

microgravity on the differentiation of EPCs pre-exposed to simulated microgravity. This

report also studied the downstream function of the differentiated cells in terms of their

migration and proliferative capabilities, release of soluble factors such as PGE2 and

NO, and ability to regulate clotting mechanisms in the body. While both the migration

and proliferation of cells were inhibited by microgravity exposure, prolonged exposure of

EPCs to microgravity resulted in decrease in the migration abilities of EPC derived EC

and the proliferative capacity of the ECs seemed to recover on longer time of exposure.

The initial decrease and subsequent increase in release of soluble factors such as NO

and PGE2 reaffirmed the possibility of recovery mechanisms being involved on

prolonged exposure to weightlessness. Finally, the assessment of whole blood and

plasma clotting, indicated that microgravity pre exposure at the progenitor stage caused

inhibition, in the ability of PE-like cells to delay blood or plasma clotting while this effect

was again nulled with increase in time of exposure of EPCs to microgravity. This data

collectively provides some insight into the role of short term exposure to low shear

stress environment due to simulated microgravity, on cardiovascular deconditioning

during space travel. Especially, the effect of microgravity on anti-thrombogenic

properties of the endothelium has not been reported before. Encouraged by the results,

the study was further extended to include human blood progenitor derived endothelial

cells. The goal was to investigate and draw comparisons between the effect of

Page 177: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

177

simulated microgravity on vascular regeneration mechanism of EPCs and ECs when

exposed prior and post differentiation respectively.

This was demonstrated in Chapter 7. The results from the study show that the

populations of human blood EPC derived ECs when exposed to simulated microgravity

proliferate faster, release higher amounts of soluble NO and lower amounts of pro-

inflammatory cytokines such as IL-6, when compared to their normal gravity cultured

controls. This can be possibly attributed to the ability of the cells to sense simulated

microgravity as a “stressful” condition and adapt by upregulating gene expression of

hsp70. However, these secretory as well as proliferative functions had no effect on the

anti-thrombogenic functions of these cells. Their anti-coagulative and anti-thrombogenic

ability as assessed by both upregulation of tPA as well their ability to delay plasma

clotting, were impaired upon exposure to simulated microgravity.

Overall this study serves as the first step towards understanding the ability of

these repair cells to regulate clotting mechanisms and cardiovascular health in

response to macroscale cues provided to them by altered growth conditions. Further

studies to assess alternate and synergistic mechanisms involved in regulation of

cardiovascular health as well as inflammatory dysfunction under microgravity are

required to develop possible cell based countermeasures to prevent cardiovascular

deconditioning due to space travel. This approach also has the potential to be applied to

develop strategies for vascular regeneration on earth using EPCs.

Conclusions

As Biomaterial Engineers, the most important toolkit at our disposal is the

structure property relationship of materials. It is this characteristic of materials that

enables flexibility in design and development of novel biomaterials that can elicit

Page 178: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

178

appropriate responses from cell systems. Given the complexity of living tissues,

techniques for tissue engineering require the use of novel external cues to aid the

process. The overall aim of this dissertation work was to study multiscale cues that can

be presented to cells and the response elicited by them and their relevance in a

vascular setting. The underlying theme of studying these interactions is to repair,

preserve and regenerate vascular tissue to alleviate conditions affecting the health of

the vascular network. The nano, meso and macro scale stimuli discussed in the above

chapters cause a unique response in a simulated vascular environment. Although this

work is not a comprehensive, all-encompassing study of all cell-material interaction

scenarios, these efforts contribute towards widening the knowledge base in this field.

This work also opens potential avenues to use these systems either separately or in

combination to develop a novel strategy for vascular tissue engineering. Therefore, this

work is a valuable addition to the field of cardiovascular tissue engineering and

regenerative therapy.

Future Work

Each chapter describes a specific and novel system of interactions at cell-

biomaterial interface. These studies represent preliminary work on characterizing the

initial response of cellular systems to cues at different scales. These results can be

further expanded to in vivo settings through animal studies and applied to organotypic

models. For example, the development of an agonist aptamer assembly for VEGFR2

receptors in endothelial cells as described in Chapter 3 opens up avenues to further

extend this work to other receptors and cell types for applications in tissue engineering,

stem cell engineering, cancer therapy etc.

Page 179: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

179

Further the information learnt from these strategies can be combined to present

multiple cues at the same time. Such strategies have been used while designing

bioactive scaffolds for vascular tissue engineering in the past46,58,98,99. One possible

avenue in this direction would be the use of novel bioactive scaffold systems based on

the Collagen POC Elastin system decorated with the VEGFR2 aptamer assembly to

capture and recruit endothelial progenitor cells from circulating blood.

Therefore, future work based on the results presented in this dissertation will

potentially advance vascular tissue engineering and help further clinical care for these

complications.

Page 180: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

180

APPENDIX PROTEIN CROSSLINKING USING CINNAMALDEHYDE VAPORS

Chapter 4 described the negative effects of glutaraldehyde crosslinking and its

potential toxicity on cells. The goal of this study is to investigate the use of

cinnamaldehyde (CA) as a relatively non-toxic cross linker to stabilize protein scaffolds.

Tissue engineering scaffolds made of proteins from animal and plant sources have

been used effectively for a variety of applications185. However, these scaffolds must be

crosslinked to stabilize the proteins before they can be used in in vitro applications.

Protein scaffolds, without proper crosslinking have inferior resistance to degradation

and mechanical strength. Therefore, to improve their physical properties, many

crosslinking strategies have been utilized such as chemical cross linkers, plasticizers as

well as ionizing radiations. Currently, the most robust and popular cross linking agents

employed are primarily aldehydes such as formaldehyde and glutaraldehyde which can

cause significant toxicity to cells unless appropriate precautions are taken186–188.

Therefore, there is a constant search for alternate natural cross-linking agents which do

not cause significant toxicity to tissues.

Cinnamaldehyde occurs naturally as an aromatic α,β-unsaturated aldehyde. It is

derived from cinnamon which is used popularly as a spice. It also has medicinal

properties and has been used as a home remedy for ailments for common cold and

digestive disorders. Cinnamon oil has also been used time and again as a food

preservative because of its excellent anti-microbial properties327. More recently it has

been identified as a crosslinking agent for protein films in the food packaging industry.

Another recent report also explores its use to cross link protein hydrogels for wound

healing applications327,328.

Page 181: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

181

This preliminary study investigates the use of cinnamaldehyde vapors for cross

linking proteins. In this work, the crosslinking action of cinnamaldehyde liquid is

compared with that of cinnamaldehyde vapors. The use of the vapor form of

cinnamaldehyde for crosslinking applications has not been reported before.

Electrospinning Gelatin Nanofibers

12.5 wt. % solution of gelatin in Tetra fluoro ethylene was prepared for

electrospinning. The mixture was vortexed for 15 min and allowed to dissolve overnight

to thoroughly mix the constituents. The gelatin solution was fed through a 25G needle at

0.8 mL/ h with a syringe pump and a voltage potential of 22-25 kV was used; the

working distance was set at 25 cm.

Cinnamaldehyde Crosslinking and Preliminary Stabilization Assessment

The gelatin scaffolds in their as spun state were soluble in water. The scaffolds

were then cross linked using the conditions mentioned in the table below. Further the

stability of the scaffolds was preliminarily tested by assessing their ability to resist

dissolution in water.

Table A-1. CA cross-linking conditions and resistance to degradation

Conditions Water stability

Un-crosslinked control Dissolved in water

Liquid CA crosslinking for 24 h at RT Stable in water

Vapor CA crosslinking for 4 days at 45⁰ C Dissolved in water

Vapor CA crosslinking for 7days at 45⁰ C Stable in water

Thermal crosslinking control for 7 days at 45⁰ C Dissolved in water

From preliminary testing, it was shown that only two conditions stabilized the

gelatin fibers, viz, the fibers crosslinked with liquid CA overnight and the CA vapor

crosslinked fibers for 1 week.

Page 182: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

182

FTIR Characterization

The crosslinking of scaffolds was confirmed by FTIR. Spectrochemical analysis

of the electrospun scaffolds crosslinked at various conditions was performed using

attenuated total reflectance Fourier transformation infrared spectroscopy (ATR-FTIR).

ATR-FTIR spectra was obtained for each of the samples using the Nicolet 6700 FTIR

Spectrometer (Thermo Scientific) and a diamond tip window. The spectra were read

over a range of 600-4000 cm-1 for each of the spectra and a total of 32 scans were used

with a resolution of 4 cm-1.

Figure A-1. FTIR spectra of cross linked fibers

FTIR analysis as shown above confirmed the crosslinking of scaffolds as

indicated by a characteristic Amide II (NH) bending vibrations peak observed at around

1550 cm-1. Additionally, Amide I (C=O stretching) and Amide A (N-H stretching)

signatures were also seen respectively at 1632- 1664 and 3320-3340 cm-1for all

scaffolds tested.

Page 183: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

183

SEM Characterization

The stable samples were washed and frozen at -80˚C for 18 h before being

lyophilized. The dried scaffolds were then mounted for SEM analysis and the structure

of the scaffolds were observed using a table top SEM (PhenomWorld ProX, Eindhoven,

Netherlands).

Figure A-2. SEM characterization of CA crosslinking. (A) Un crosslinked gelatin fibers (B) Liquid CA crosslinked fibers after 24 h at room temperature (C) CA vapor

crosslinked fibers after 7 days at 45⁰C. All images are at a magnification of

5000 X.

The preliminary results from this study indicates CA vapors are effective in cross

linking protein fibers. FTIR spectra confirm this hypothesis. The SEM images

interestingly indicate that there is coalescing of fibers and the morphology changes on

cross linking. Further comprehensive evaluation of the crosslinked fibers as well as their

structure is required to further develop strategy to create functional scaffolds.

A B C

Page 184: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

184

LIST OF REFERENCES

1. WHO | Cardiovascular diseases (CVDs). WHO. 2016. http://www.who.int/mediacentre/factsheets/fs317/en/. Accessed April 26, 2017.

2. WHO | Global status report on noncommunicable diseases 2014. http://www.who.int/nmh/publications/ncd-status-report-2014/en/. Accessed July 2, 2015.

3. Roger VL, Go AS, Lloyd-Jones DM, et al. Heart disease and stroke statistics--2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2-e220. doi:10.1161/CIR.0b013e31823ac046.

4. Lam MT, Wu JC. Biomaterial applications in cardiovascular tissue repair and regeneration. Expert Rev Cardiovasc Ther. 2012;10(8):1039-1049. doi:10.1586/erc.12.99.

5. Cardiovascular Medicine. Springer Science & Business Media; 2007. https://books.google.com/books?id=CVdGAAAAQBAJ&pgis=1. Accessed July 5, 2015.

6. Nerem RM, Seliktar D. Vascular Tissue Engineering. Annu Rev Biomed Eng. 2001;3(1):225-243. doi:10.1146/annurev.bioeng.3.1.225.

7. Ravi S, Chaikof EL. Biomaterials for vascular tissue engineering. Regen Med. 2010;5(1):107-120. doi:10.2217/rme.09.77.

8. Catto V, Farè S, Freddi G, Tanzi MC. Vascular Tissue Engineering: Recent Advances in Small Diameter Blood Vessel Regeneration. ISRN Vasc Med. 2014;2014:1-27. doi:10.1155/2014/923030.

9. Teebken OE, Haverich A. Tissue engineering of small diameter vascular grafts. Eur J Vasc Endovasc Surg. 2002;23(6):475-485. doi:10.1053/ejvs.2002.1654.

10. Berzat A, Hall A. Cellular responses to extracellular guidance cues. EMBO J. 2010;29(16):2734-2745. doi:10.1038/emboj.2010.170.

11. Rice JJ, Martino MM, De Laporte L, Tortelli F, Briquez PS, Hubbell JA. Engineering the Regenerative Microenvironment with Biomaterials. Adv Healthc Mater. 2013;2(1):57-71. doi:10.1002/adhm.201200197.

12. Mashayekhan S, Hajiabbas M, Fallah A. Stem Cells in Tissue Engineering. In: Pluripotent Stem Cells. Vol InTech; 2013. doi:10.5772/54371.

13. Paratore C, Hagedorn L, Floris J, et al. Cell-intrinsic and cell-extrinsic cues regulating lineage decisions in multipotent neural crest-derived progenitor cells. Int J Dev Biol. 2002;46(1):193-200. http://www.ncbi.nlm.nih.gov/pubmed/11902683. Accessed April 26, 2017.

Page 185: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

185

14. Gilchrist CL, Ruch DS, Little D, Guilak F. Micro-scale and meso-scale architectural cues cooperate and compete to direct aligned tissue formation. Biomaterials. 2014;35(38):10015-10024. doi:10.1016/j.biomaterials.2014.08.047.

15. Ratner BD (Buddy D. Biomaterials Science : An Introduction to Materials in Medicine. Elsevier Academic Press; 2004.

16. Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polym Sci. 2007;32(8-9):762-798. doi:10.1016/j.progpolymsci.2007.05.017.

17. Burdick JA, Vunjak-Novakovic G. Engineered microenvironments for controlled stem cell differentiation. Tissue Eng Part A. 2009;15(2):205-219. doi:10.1089/ten.tea.2008.0131.

18. Kim D-H, Lipke EA, Kim P, et al. Nanoscale cues regulate the structure and function of macroscopic cardiac tissue constructs. Proc Natl Acad Sci U S A. 2010;107(2):565-570. doi:10.1073/pnas.0906504107.

19. Go AS, Mozaffarian D, Roger VL, et al. Heart disease and stroke statistics--2014 update: a report from the American Heart Association. Circulation. 2014;129(3):e28-e292. doi:10.1161/01.cir.0000441139.02102.80.

20. Lloyd-Jones D, Adams RJ, Brown TM, et al. Heart Disease and Stroke Statistics—2010 Update A Report From the American Heart Association. Circulation. 2010;121(7). doi:10.1161/CIRCULATIONAHA.109.192667.

21. ROSE GA, BLACKBURN H, GILLUM RF, PRINEAS RJ. Cardiovascular survey methods. Monogr Ser World Heal Organ. 1982;56(2nd edition). https://www.cabdirect.org/cabdirect/abstract/19832701107. Accessed May 29, 2017.

22. Vesely I. Heart Valve Tissue Engineering. Circ Res. 2005;97(8). http://circres.ahajournals.org/content/97/8/743.short. Accessed May 29, 2017.

23. Stock UA, Vacanti JP. Tissue Engineering: Current State and Prospects. Annu Rev Med. 2001;52(1):443-451. doi:10.1146/annurev.med.52.1.443.

24. Bouten CVC, Dankers PYW, Driessen-Mol A, Pedron S, Brizard AMA, Baaijens FPT. Substrates for cardiovascular tissue engineering. Adv Drug Deliv Rev. 2011;63(4):221-241. doi:10.1016/j.addr.2011.01.007.

25. Nugent HM, Edelman ER. Tissue Engineering Therapy for Cardiovascular Disease. Circ Res. 2003;92(10):1068-1078. doi:10.1161/01.RES.0000073844.41372.38.

26. Stevens MM. Exploring and Engineering the Cell Surface Interface. Science (80- ). 2005;310(5751):1135-1138. doi:10.1126/science.1106587.

Page 186: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

186

27. Gasiorowski JZ, Murphy CJ, Nealey PF. Biophysical Cues and Cell Behavior: The Big Impact of Little Things. Annu Rev Biomed Eng. 2013;15(1):155-176. doi:10.1146/annurev-bioeng-071811-150021.

28. Feynman R. There’s plenty of room at the bottom. Eng Sci. 1960;23(5):22-36. doi:10.1016/j.applthermaleng.2013.04.038.

29. Dvir T, Timko BP, Kohane DS, Langer R. Nanotechnological strategies for engineering complex tissues. Nat Nanotechnol. 2011;6(1):13-22. doi:10.1038/nnano.2010.246.

30. Di Cio S, Gautrot JE. Cell sensing of physical properties at the nanoscale: Mechanisms and control of cell adhesion and phenotype. Acta Biomater. 2016;30:26-48. doi:10.1016/j.actbio.2015.11.027.

31. Martino MM, Brkic S, Bovo E, et al. Extracellular Matrix and Growth Factor Engineering for Controlled Angiogenesis in Regenerative Medicine. Front Bioeng Biotechnol. 2015;3:45. doi:10.3389/fbioe.2015.00045.

32. Dvir T, Timko BP, Kohane DS, Langer R. Nanotechnological strategies for engineering complex tissues. Nat Publ Gr. 2010;6. doi:10.1038/nnano.2010.246.

33. Jones JR. Observing cell response to biomaterials. Mater Today. 2006;9(12):34-43. doi:10.1016/S1369-7021(06)71741-2.

34. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general approaches and a review of recent developments. J R Soc Interface. 2011;8(55):153-170. doi:10.1098/rsif.2010.0223.

35. Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346(6287):818-822. doi:10.1038/346818a0.

36. Morris KN, Jensen KB, Julin CM, Weil M, Gold L. High affinity ligands from in vitro selection: Complex targets. Biochemistry. 1998;95:2902-2907. http://www.pnas.org/content/95/6/2902.full.pdf. Accessed April 30, 2017.

37. Simón-Yarza T, Formiga FR, Tamayo E, Pelacho B, Prosper F, Blanco-Prieto MJ. Vascular endothelial growth factor-delivery systems for cardiac repair: an overview. Theranostics. 2012;2(6):541-552. doi:10.7150/thno.3682.

38. Simons M, Annex BH, Laham RJ, et al. Pharmacological Treatment of Coronary Artery Disease With Recombinant Fibroblast Growth Factor-2. Circulation. 2002;105(7). http://circ.ahajournals.org/content/105/7/788.short. Accessed May 29, 2017.

39. Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669-676. doi:10.1038/nm0603-669.

Page 187: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

187

40. Govender S, Csimma C, Genant HK, et al. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg Am. 2002;84-A(12):2123-2134. http://www.ncbi.nlm.nih.gov/pubmed/12473698. Accessed May 29, 2017.

41. Vaccaro AR, Whang PG, Patel T, et al. The safety and efficacy of OP-1 (rhBMP-7) as a replacement for iliac crest autograft for posterolateral lumbar arthrodesis: minimum 4-year follow-up of a pilot study. Spine J. 2008;8(3):457-465. doi:10.1016/j.spinee.2007.03.012.

42. Henry TD, Annex BH, McKendall GR, et al. The VIVA Trial. Circulation. 2003;107(10). http://circ.ahajournals.org/content/107/10/1359.short. Accessed May 29, 2017.

43. Schwarz ER, Speakman MT, Patterson M, et al. Evaluation of the effects of intramyocardial injection of DNA expressing vascular endothelial growth factor (VEGF) in a myocardial infarction model in the rat—angiogenesis and angioma formation. J Am Coll Cardiol. 2000;35(5):1323-1330. doi:10.1016/S0735-1097(00)00522-2.

44. Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18(14):3964-3972. doi:10.1093/emboj/18.14.3964.

45. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general approaches and a review of recent developments. J R Soc Interface. 2010;8(55). http://rsif.royalsocietypublishing.org/content/8/55/153.short. Accessed May 20, 2017.

46. Schleicher M, Hansmann J, Kluger P, et al. In vivo endothelialization of heart valves using high specific DNA-aptamers as capture molecules for circulating endothelial progenitor cells (EPCs). Thorac Cardiovasc Surg. 2010;58(S 01). doi:10.1055/s-0029-1246794.

47. Keefe AD, Pai S, Ellington A. Aptamers as therapeutics. Nat Rev Drug Discov. 2010;9(7):537-550. doi:10.1038/nrd3141.

48. Jayasena SD. Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin Chem. 1999;45(9):1628-1650. http://www.ncbi.nlm.nih.gov/pubmed/10471678. Accessed May 29, 2017.

49. Gold L, Singer B, He YY, Brody E. SELEX and the evolution of genomes. Curr Opin Genet Dev. 1997;7(6):848-851. doi:10.1016/S0959-437X(97)80050-0.

50. Xiao Z, Shangguan D, Cao Z, Fang X, Tan W. Cell-Specific Internalization Study of an Aptamer from Whole Cell Selection. Chem - A Eur J. 2008;14(6):1769-1775. doi:10.1002/chem.200701330.

Page 188: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

188

51. Hermann T, Patel DJ. Adaptive Recognition by Nucleic Acid Aptamers. Science (80- ). 2000;287(5454). http://science.sciencemag.org/content/287/5454/820. Accessed April 30, 2017.

52. Guo K-T, Schäfer R, Paul A, Ziemer G, Wendel HP. Aptamer-based strategies for stem cell research. Mini Rev Med Chem. 2007;7(7):701-705. http://www.ncbi.nlm.nih.gov/pubmed/17627582. Accessed May 29, 2017.

53. Brody EN, Gold L. Aptamers as therapeutic and diagnostic agents. J Biotechnol. 2000;74(1):5-13. http://www.ncbi.nlm.nih.gov/pubmed/10943568. Accessed April 30, 2017.

54. Song K-M, Lee S, Ban C. Aptamers and Their Biological Applications. Sensors. 2012;12(12):612-631. doi:10.3390/s120100612.

55. Brody EN, Gold L. Aptamers as therapeutic and diagnostic agents. J Biotechnol. 2000;74(1):5-13. http://www.ncbi.nlm.nih.gov/pubmed/10943568. Accessed May 29, 2017.

56. Ng EWM, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 2006;5(2):123-132. doi:10.1038/nrd1955.

57. Zhang K, Sefah K, Tang L, et al. A Novel Aptamer Developed for Breast Cancer Cell Internalization. ChemMedChem. 2012;7(1):79-84. doi:10.1002/cmdc.201100457.

58. Guo J, Gao X, Su L, et al. Aptamer-functionalized PEG-PLGA nanoparticles for enhanced anti-glioma drug delivery. Biomaterials. 2011;32(31):8010-8020. doi:10.1016/j.biomaterials.2011.07.004.

59. Hermann N, Brorson M, Topsøe H. N. Hermann, M. Brorson, H. Topsoe, Catalysis Letters, 65 (2000) 169-174. Catal Letters. 2000;65:169-174.

60. Ni X, Castanares M, Mukherjee A, Lupold SE. Nucleic acid aptamers: clinical applications and promising new horizons. Curr Med Chem. 2011;18(27):4206-4214. http://www.ncbi.nlm.nih.gov/pubmed/21838685. Accessed May 29, 2017.

61. Dollins CM, Nair S, Boczkowski D, et al. Assembling OX40 aptamers on a molecular scaffold to create a receptor-activating aptamer. Chem Biol. 2008;15(7):675-682. doi:10.1016/j.chembiol.2008.05.016.

62. Iaboni M, Fontanella R, Rienzo A, et al. Targeting Insulin Receptor with a Novel Internalizing Aptamer. Mol Ther Nucleic Acids. 2016;5(9):e365. doi:10.1038/mtna.2016.73.

63. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010;123(24). http://jcs.biologists.org/content/123/24/4195. Accessed May

Page 189: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

189

29, 2017.

64. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix Elasticity Directs Stem Cell Lineage Specification. Cell. 2006;126(4):677-689. doi:10.1016/j.cell.2006.06.044.

65. Wells RG. The role of matrix stiffness in regulating cell behavior. Hepatology. 2008;47(4):1394-1400. doi:10.1002/hep.22193.

66. Lam MT, Wu JC. Biomaterial applications in cardiovascular tissue repair and regeneration. Expert Rev Cardiovasc Ther. 2012;10(8):1039-1049. doi:10.1586/erc.12.99.

67. Jaganathan SK, Supriyanto E, Murugesan S, Balaji A, Asokan MK. Biomaterials in cardiovascular research: applications and clinical implications. Biomed Res Int. 2014;2014:459465. doi:10.1155/2014/459465.

68. Li M, Mondrinos MJ, Gandhi MR, Ko FK, Weiss AS, Lelkes PI. Electrospun protein fibers as matrices for tissue engineering. Biomaterials. 2005;26(30):5999-6008. doi:10.1016/j.biomaterials.2005.03.030.

69. Ruso J, Messina P. Biopolymers in Regenerative Medicine: Overview, Current Advances and Future Trends. In: Biopolymers for Medical Applications. Vol Taylor & Francis Group, 6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487-2742: CRC Press; 2016:1-37. doi:10.1201/9781315368863-2.

70. Ulery BD, Nair LS, Laurencin CT. Biomedical applications of biodegradable polymers. J Polym Sci Part B Polym Phys. 2011;49(12):832-864. doi:10.1002/polb.22259.

71. Gunatillake P, Mayadunne R, Adhikari R. Recent developments in biodegradable synthetic polymers. Biotechnol Annu Rev. 2006;12:301-347. doi:10.1016/S1387-2656(06)12009-8.

72. Teebken OE, Haverich A. Tissue Engineering of Small Diameter Vascular Grafts. Eur J Vasc Endovasc Surg. 2002;23(6):475-485. doi:10.1053/ejvs.2002.1654.

73. De BAKEY ME. The Fate of Dacron Vascular Grafts. Arch Surg. 1964;89(5):755. doi:10.1001/archsurg.1964.01320050001001.

74. Leitão AF, Silva I, Faria M, Gama M. Vascular Grafts: Polymeric Materials. In: Encyclopedia of Biomedical Polymers and Polymeric Biomaterials. Vol Taylor & Francis; 2016:8144-8164. doi:10.1081/E-EBPP-120050697.

75. Gorbet MB, Sefton M V. Biomaterial-associated thrombosis: roles of coagulation factors, complement, platelets and leukocytes. Biomaterials. 2004;25(26):5681-5703. doi:10.1016/j.biomaterials.2004.01.023.

76. Sayers RD, Raptis S, Berce M, Miller JH. Long-term results of femorotibial bypass

Page 190: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

190

with vein or polytetrafluoroethylene. Br J Surg. 1998;85(7):934-938. doi:10.1046/j.1365-2168.1998.00765.x.

77. Gunatillake PA, Adhikari R, Gadegaard N. Biodegradable synthetic polymers for tissue engineering. Eur Cells Mater. 2003;5:1-16. doi:vol005a01 [pii].

78. Weinberg CB, Bell E. A blood vessel model constructed from collagen and cultured vascular cells. Science. 1986;231(4736):397-400. http://www.ncbi.nlm.nih.gov/pubmed/2934816. Accessed May 29, 2017.

79. Boland ED, Matthews JA, Pawlowski KJ, Simpson DG, Wnek GE, Bowlin GL. Electrospinning collagen and elastin: preliminary vascular tissue engineering. Front Biosci. 2004;9:1422-1432. doi:10.2741/1313.

80. Prabhakaran MP, Nair AS, Kai D, Ramakrishna S. Electrospun composite scaffolds containing poly(octanediol-co-citrate) for cardiac tissue engineering. Biopolymers. 2012;97(7):529-538. doi:10.1002/bip.22035.

81. Liu J, Argenta L, Morykwas M, Wagner WD. Properties of single electrospun poly (diol citrate)-collagen-proteoglycan nanofibers for arterial repair and in applications requiring viscoelasticity. J Biomater Appl. 2014;28(5):729-738. doi:10.1177/0885328213477893.

82. Yang J, Webb AR, Ameer GA. Novel Citric Acid-Based Biodegradable Elastomers for Tissue Engineering. Adv Mater. 2004;16(6):511-516. doi:10.1002/adma.200306264.

83. Yang J, Webb AR, Pickerill SJ, Hageman G, Ameer GA. Synthesis and evaluation of poly(diol citrate) biodegradable elastomers. Biomaterials. 2006;27(9):1889-1898. doi:10.1016/j.biomaterials.2005.05.106.

84. Motlagh D, Allen J, Hoshi R, Yang J, Lui K, Ameer G. Hemocompatibility evaluation of poly(diol citrate) in vitro for vascular tissue engineering. J Biomed Mater Res A. 2007;82(4):907-916. doi:10.1002/jbm.a.31211.

85. Allen J, Khan S, Serrano MC, Ameer G. Characterization of porcine circulating progenitor cells: toward a functional endothelium. Tissue Eng Part A. 2008;14(1):183-194. doi:10.1089/ten.a.2007.0265.

86. Seifu DG, Purnama A, Mequanint K, Mantovani D. Small-diameter vascular tissue engineering. Nat Rev Cardiol. 2013;10(7):410-421. doi:10.1038/nrcardio.2013.77.

87. Wang X, Lin P, Yao Q, Chen C. Development of small-diameter vascular grafts. World J Surg. 2007;31(4):682-689. doi:10.1007/s00268-006-0731-z.

88. Griffith LG, Naughton G. Tissue Engineering--Current Challenges and Expanding Opportunities. Science (80- ). 2002;295(5557). http://science.sciencemag.org/content/295/5557/1009. Accessed May 29, 2017.

Page 191: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

191

89. Rnjak-Kovacina J, Wise SG, Li Z, et al. Tailoring the porosity and pore size of electrospun synthetic human elastin scaffolds for dermal tissue engineering. Biomaterials. 2011;32(28):6729-6736. doi:10.1016/j.biomaterials.2011.05.065.

90. McClure MJ, Sell SA, Simpson DG, Walpoth BH, Bowlin GL. A three-layered electrospun matrix to mimic native arterial architecture using polycaprolactone, elastin, and collagen: A preliminary study. Acta Biomater. 2010;6(7):2422-2433. doi:10.1016/j.actbio.2009.12.029.

91. Hansson GK. Inflammation, Atherosclerosis, and Coronary Artery Disease. N Engl J Med. 2005;352(16):1685-1695. doi:10.1056/NEJMra043430.

92. Hill JM, Zalos G, Halcox JPJ, et al. Circulating Endothelial Progenitor Cells, Vascular Function, and Cardiovascular Risk. N Engl J Med. 2003;348(7):593-600. doi:10.1056/NEJMoa022287.

93. McClure MJ, Simpson DG, Bowlin GL. Tri-layered vascular grafts composed of polycaprolactone, elastin, collagen, and silk: Optimization of graft properties. J Mech Behav Biomed Mater. 2012;10:48-61. doi:10.1016/j.jmbbm.2012.02.026.

94. Madurantakam PA, Rodriguez IA, Garg K, McCool JM, Moon PC, Bowlin GL. Compression of Multilayered Composite Electrospun Scaffolds: A Novel Strategy to Rapidly Enhance Mechanical Properties and Three Dimensionality of Bone Scaffolds. Adv Mater Sci Eng. 2013;2013:1-9. doi:10.1155/2013/561273.

95. Miyazu K, Kawahara D, Ohtake H, Watanabe G, Matsuda T. Luminal surface design of electrospun small-diameter graft aiming at in situ capture of endothelial progenitor cell. J Biomed Mater Res - Part B Appl Biomater. 2010;94(1):53-63. doi:10.1002/jbm.b.31623.

96. Ercolani E, Del Gaudio C, Bianco A. Vascular tissue engineering of small-diameter blood vessels: reviewing the electrospinning approach. J Tissue Eng Regen Med. 2015;9(8):861-888. doi:10.1002/term.1697.

97. Rocco KA, Maxfield MW, Best CA, Dean EW, Breuer CK. In vivo applications of electrospun tissue-engineered vascular grafts: a review. Tissue Eng Part B Rev. 2014;20(6):628-640. doi:10.1089/ten.TEB.2014.0123.

98. Wendel HP, Avci-Adali M, Ziemer G. Endothelial progenitor cell capture stents--hype or hope? Int J Cardiol. 2010;145(1):115-117; author reply 117-118. doi:10.1016/j.ijcard.2009.06.020.

99. Hoffmann J, Paul A, Harwardt M, et al. Immobilized DNA aptamers used as potent attractors for porcine endothelial precursor cells. J Biomed Mater Res A. 2008;84(3):614-621. doi:10.1002/jbm.a.31309.

100. Edfors F, Boström T, Forsström B, et al. Immunoproteomics using polyclonal antibodies and stable isotope-labeled affinity-purified recombinant proteins. Mol

Page 192: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

192

Cell Proteomics. 2014;13(6):1611-1624. doi:10.1074/mcp.M113.034140.

101. Chen Q, Thouas GA. Metallic implant biomaterials. Mater Sci Eng R Reports. 2015;87:1-57. doi:10.1016/j.mser.2014.10.001.

102. Mani G, Feldman MD, Patel D, Agrawal CM. Coronary stents: A materials perspective. Biomaterials. 2007;28(9):1689-1710. doi:10.1016/j.biomaterials.2006.11.042.

103. Hermawan H, Dubé D, Mantovani D. Developments in metallic biodegradable stents. Acta Biomater. 2010;6(5):1693-1697. doi:10.1016/j.actbio.2009.10.006.

104. Ruygrok PN, Serruys PW. Intracoronary Stenting. Circulation. 1996;94(5).

105. Erne P, Schier M, Resink TJ. The Road to Bioabsorbable Stents: Reaching Clinical Reality? Cardiovasc Intervent Radiol. 2006;29(1):11-16. doi:10.1007/s00270-004-0341-9.

106. Hermawan H, Dubé D, Mantovani D. Developments in metallic biodegradable stents. Acta Biomater. 2010;6(5):1693-1697. doi:10.1016/j.actbio.2009.10.006.

107. Moravej M, Mantovani D. Biodegradable Metals for Cardiovascular Stent Application: Interests and New Opportunities. Int J Mol Sci. 2011;12(12):4250-4270. doi:10.3390/ijms12074250.

108. Waksman R, Pakala R. Biodegradable and bioabsorbable stents. Curr Pharm Des. 2010;16(36):4041-4051. http://www.ncbi.nlm.nih.gov/pubmed/21208182. Accessed February 20, 2017.

109. Shen C, Liu X, Fan B, et al. Mechanical properties, in vitro degradation behavior, hemocompatibility and cytotoxicity evaluation of Zn–1.2Mg alloy for biodegradable implants. RSC Adv. 2016;6(89):86410-86419. doi:10.1039/C6RA14300H.

110. De Waha A, Stefanini GG, King LA, et al. Long-term outcomes of biodegradable polymer versus durable polymer drug-eluting stents in patients with diabetes a pooled analysis of individual patient data from 3 randomized trials. Int J Cardiol. 2013;168(6):5162-5166. doi:10.1016/j.ijcard.2013.07.263.

111. Zeng R, Dietzel W, Witte F, Hort N, Blawert C. Progress and Challenge for Magnesium Alloys as Biomaterials. Adv Eng Mater. 2008;10(8):B3-B14. doi:10.1002/adem.200800035.

112. Stanciu L, Heiden M, Walker E. Biotechnology &amp; Biomaterials Magnesium, Iron and Zinc Alloys, the Trifecta of Bioresorbable Orthopaedic and Vascular Implantation -A Review. J Biotechnol Biomater. 2015;5(5). doi:10.4172/2155-952X.1000178.

113. Rukshin V, Azarbal B, Shah PK, et al. Intravenous Magnesium in Experimental

Page 193: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

193

Stent Thrombosis in Swine. Arterioscler Thromb Vasc Biol. 2001;21(9):1544-1549.

114. Shechter M. Does Magnesium Have a Role in the Treatment of Patients with Coronary Artery Disease? Am J Cardiovasc Drugs. 2003;3(4):231-239. doi:10.2165/00129784-200303040-00001.

115. Eric Jones J, Chen M, Yu Q. Corrosion resistance improvement for 316L stainless steel coronary artery stents by trimethylsilane plasma nanocoatings. J Biomed Mater Res Part B Appl Biomater. 2014;102(7):1363-1374. doi:10.1002/jbm.b.33115.

116. Alsberg E, von Recum HA, Mahoney MJ. Environmental cues to guide stem cell fate decision for tissue engineering applications. Expert Opin Biol Ther. 2006;6(9):847-866. doi:10.1517/14712598.6.9.847.

117. Dahl SLM, Kypson AP, Lawson JH, et al. Readily Available Tissue-Engineered Vascular Grafts. Sci Transl Med. 2011;3(68):68ra9-ra68ra9. doi:10.1126/scitranslmed.3001426.

118. Hansmann J, Groeber F, Kahlig A, Kleinhans C, Walles H. Bioreactors in tissue engineering-principles, applications and commercial constraints. Biotechnol J. 2013;8(3):298-307. doi:10.1002/biot.201200162.

119. Klaus DM. Clinostats and bioreactors. Gravit Space Biol Bull. 2001;14(2):55-64. http://www.ncbi.nlm.nih.gov/pubmed/11865869. Accessed August 31, 2015.

120. Barron V, Lyons E, Stenson-Cox C, McHugh PE, Pandit a. Bioreactors for cardiovascular cell and tissue growth: a review. Ann Biomed Eng. 2003;31(9):1017-1030. doi:10.1114/1.1603260.

121. Pörtner R, Nagel-Heyer S, Goepfert C, Adamietz P, Meenen NM. Bioreactor design for tissue engineering. J Biosci Bioeng. 2005;100(3):235-245. doi:10.1263/jbb.100.235.

122. Pörtner R, Giese C. An Overview on Bioreactor Design, Prototyping and Process Control for Reproducible Three-Dimensional Tissue Culture. In: Drug Testing in Vitro. Vol Weinheim, Germany: Wiley-VCH Verlag GmbH & Co. KGaA; 2006:53-78. doi:10.1002/9783527609611.ch2.

123. Martin I, Wendt D, Heberer M. The role of bioreactors in tissue engineering. Trends Biotechnol. 2004;22(2):80-86. doi:10.1016/j.tibtech.2003.12.001.

124. Pei M, Solchaga LA, Seidel J, et al. Bioreactors mediate the effectiveness of tissue engineering scaffolds. FASEB J. 2002;16(12):1691-1694. doi:10.1096/fj.02-0083fje.

125. Freed LE, Guilak F, Guo XE, et al. Advanced Tools for Tissue Engineering:

Page 194: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

194

Scaffolds, Bioreactors, and Signaling. Tissue Eng. 2006;12(12):3285-3305. doi:10.1089/ten.2006.12.3285.

126. Unsworth BR, Lelkes PI. Growing tissues in microgravity. Nat Med. 1998;4(8):901-907. doi:10.1038/nm0898-901.

127. Hammond TG, Hammond JM. Optimized suspension culture: the rotating-wall vessel. Am J Physiol Ren Physiol. 2001;281(1):F12-F25. http://ajprenal.physiology.org/content/281/1/F12.full.pdf+html. Accessed November 2, 2015.

128. Cogoli A. Microgravity and mammalian cells. Cell Differ Dev. 1989;27:180.

129. Grimm D, Wehland M, Pietsch J, et al. Growing tissues in real and simulated microgravity: new methods for tissue engineering. Tissue Eng Part B Rev. 2014;20(6):555-566. doi:10.1089/ten.TEB.2013.0704.

130. Blaber E, Sato K, Almeida EAC. Stem cell health and tissue regeneration in microgravity. Stem Cells Dev. 2014;23 Suppl 1:73-78. doi:10.1089/scd.2014.0408.

131. Nickerson CA, Pellis NR (Neal R, Ott CM. Effect of Spaceflight and Spaceflight Analogue Culture on Human and Microbial Cells : Novel Insights into Disease Mechanisms.

132. Barrila J, Wilson JW, Soni A, Yang J, Mark Ott C, Nickerson CA. Using Spaceflight and Spaceflight Analogue Culture for Novel Mechanistic Insight into Salmonella Pathogenesis. In: Effect of Spaceflight and Spaceflight Analogue Culture on Human and Microbial Cells. Vol New York, NY: Springer New York; 2016:209-235. doi:10.1007/978-1-4939-3277-1_11.

133. Cotrupi S, Ranzani D, Maier JAM. Impact of modeled microgravity on microvascular endothelial cells. Biochim Biophys Acta. 2005;1746(2):163-168. doi:10.1016/j.bbamcr.2005.10.002.

134. Martinez EM, Yoshida MC, Candelario TLT, Hughes-Fulford M. Spaceflight and simulated microgravity cause a significant reduction of key gene expression in early T-cell activation. Am J Physiol Regul Integr Comp Physiol. 2015;308(6):R480-R488. doi:10.1152/ajpregu.00449.2014.

135. Versari S, Longinotti G, Barenghi L, Maier JAM, Bradamante S. The challenging environment on board the International Space Station affects endothelial cell function by triggering oxidative stress through thioredoxin interacting protein overexpression: the ESA-SPHINX experiment. FASEB J. 2013;27(11):4466-4475. doi:10.1096/fj.13-229195.

136. Villa A, Versari S, Maier JAM, Bradamante S. Cell behavior in simulated microgravity: A comparison of results obtained with RWV and RPM. Gravitational

Page 195: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

195

Sp Res. 18(2):89-90. http://www.gravitationalandspacebiology.org/index.php/journal/article/view/352. Accessed November 2, 2015.

137. Freed LE, Vunjak-Novakovic G. Microgravity tissue engineering. Vitr Cell Dev Biol - Anim. 1997;33(5):381-385. doi:10.1007/s11626-997-0009-2.

138. Grimm D, Bauer J, Ulbrich C, et al. Different Responsiveness of Endothelial Cells to Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor Added to Culture Media Under Gravity and Simulated Microgravity.

139. Wehland M, Ma X, Braun M, et al. The impact of altered gravity and vibration on endothelial cells during a parabolic flight. Cell Physiol Biochem. 2013;31(2-3):432-451. doi:10.1159/000343380.

140. Chiu B, Z-M Wan J, Abley D, Akabutu J. Induction of vascular endothelial phenotype and cellular proliferation from human cord blood stem cells cultured in simulated microgravity. Acta Astronaut. 2005;56(9-12):918-922. doi:10.1016/j.actaastro.2005.01.018.

141. Hughes-Fulford M, Tjandrawinata R, Fitzgerald J, Gasuad K, Gilbertson V. Effects of microgravity on osteoblast growth. Gravit Space Biol Bull. 1998;11(2):51-60. http://www.ncbi.nlm.nih.gov/pubmed/11540639. Accessed May 29, 2017.

142. Wang R, Chu GC-Y, Zhau HE, Chung LWK. Using a Spaceflight Three-Dimensional Microenvironment to Probe Cancer–Stromal Interactions. In: Effect of Spaceflight and Spaceflight Analogue Culture on Human and Microbial Cells. Vol New York, NY: Springer New York; 2016:131-150. doi:10.1007/978-1-4939-3277-1_7.

143. Monici M, Morbidelli L, Maier JAM, Cialdai F. The Impact of Microgravity and Hypergravity on Endothelial Cells. BioMed Res Int 2015. 2015;Volume 201:13. doi:http://dx.doi.org/10.1155/2015/434803.

144. Dai K, Wang Y, Yan R, et al. Effects of microgravity and hypergravity on platelet functions. Thromb Haemost. 2009;101(5):902-910. http://www.ncbi.nlm.nih.gov/pubmed/19404544. Accessed September 8, 2015.

145. Gestwicki JE, Cairo CW, Strong LE, Oetjen KA, Kiessling LL. Influencing receptor-ligand binding mechanisms with multivalent ligand architecture. J Am Chem Soc. 2002;124(50):14922-14933. http://www.ncbi.nlm.nih.gov/pubmed/12475334. Accessed April 30, 2017.

146. Douglas MR, Morrison KC, Jacques SJ, et al. Off-target effects of epidermal growth factor receptor antagonists mediate retinal ganglion cell disinhibited axon growth. Brain. 2009;132(11):3102-3121. doi:10.1093/brain/awp240.

147. Jayasena SD. Aptamers: an emerging class of molecules that rival antibodies in

Page 196: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

196

diagnostics. Clin Chem. 1999;45(9):1628-1650. http://www.ncbi.nlm.nih.gov/pubmed/10471678. Accessed April 30, 2017.

148. Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249(4968):505-510. http://www.ncbi.nlm.nih.gov/pubmed/2200121. Accessed April 30, 2017.

149. White RR, Sullenger BA, Rusconi CP. Developing aptamers into therapeutics. J Clin Invest. 2000;106(8):929-934. doi:10.1172/JCI11325.

150. Wang C, Zhang M, Yang G, et al. Single-stranded DNA aptamers that bind differentiated but not parental cells: subtractive systematic evolution of ligands by exponential enrichment. J Biotechnol. 2003;102(1):15-22. http://www.ncbi.nlm.nih.gov/pubmed/12668310. Accessed April 30, 2017.

151. Ruckman J, Green LS, Beeson J, et al. 2’-Fluoropyrimidine RNA-based aptamers to the 165-amino acid form of vascular endothelial growth factor (VEGF165). Inhibition of receptor binding and VEGF-induced vascular permeability through interactions requiring the exon 7-encoded domain. J Biol Chem. 1998;273(32):20556-20567. http://www.ncbi.nlm.nih.gov/pubmed/9685413. Accessed April 30, 2017.

152. White RR, Shan S, Rusconi CP, et al. Inhibition of rat corneal angiogenesis by a nuclease-resistant RNA aptamer specific for angiopoietin-2. Proc Natl Acad Sci U S A. 2003;100(9):5028-5033. doi:10.1073/pnas.0831159100.

153. Ng EWM, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 2006;5(2):123-132. doi:10.1038/nrd1955.

154. Mori Y, Nagaoka S, Takiuchi H, et al. A new antithrombogenic material with long polyethyleneoxide chains. Trans Am Soc Artif Intern Organs. 1982;28:459-463. http://www.ncbi.nlm.nih.gov/pubmed/7164281. Accessed April 30, 2017.

155. Leppänen V-M, Prota AE, Jeltsch M, et al. Structural determinants of growth factor binding and specificity by VEGF receptor 2. Proc Natl Acad Sci U S A. 2010;107(6):2425-2430. doi:10.1073/pnas.0914318107.

156. Gonzaga F, Souza RD’, Brook MA. Oriented crystallization of ultra-thin (2 nm) gold nanoplatelets inside a reactive hydrophobic polymeric matrix. doi:10.1039/c0sm00743a.

157. Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990;61(2):203-212. http://www.ncbi.nlm.nih.gov/pubmed/2158859. Accessed April 30, 2017.

158. Koch S, Claesson-Welsh L. Signal Transduction by Vascular Endothelial Growth Factor Receptors. Cold Spring Harb Perspect Med. 2012;2(7):a006502-a006502.

Page 197: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

197

doi:10.1101/cshperspect.a006502.

159. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell. 2007;129(7):1261-1274. doi:10.1016/j.cell.2007.06.009.

160. Hers I, Vincent EE, Tavaré JM. Akt signalling in health and disease. Cell Signal. 2011;23(10):1515-1527. doi:10.1016/j.cellsig.2011.05.004.

161. Fujio Y, Walsh K. Akt mediates cytoprotection of endothelial cells by vascular endothelial growth factor in an anchorage-dependent manner. J Biol Chem. 1999;274(23):16349-16354. http://www.ncbi.nlm.nih.gov/pubmed/10347193. Accessed April 30, 2017.

162. Gerber HP, McMurtrey A, Kowalski J, et al. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3’-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem. 1998;273(46):30336-30343. http://www.ncbi.nlm.nih.gov/pubmed/9804796. Accessed April 30, 2017.

163. Koolwijk P, Peters E, van der Vecht B, et al. Involvement of VEGFR-2 (kdr/flk-1) but not VEGFR-1 (flt-1) in VEGF-A and VEGF-C-induced tube formation by human microvascular endothelial cells in fibrin matrices in vitro. Angiogenesis. 2001;4(1):53-60. doi:10.1023/A:1016637700638.

164. Vu MT, Smith CF, Burger PC, Klintworth GK. An evaluation of methods to quantitate the chick chorioallantoic membrane assay in angiogenesis. Lab Invest. 1985;53(4):499-508. http://www.ncbi.nlm.nih.gov/pubmed/2413278. Accessed May 2, 2017.

165. Lokman NA, Elder ASF, Ricciardelli C, Oehler MK. Chick chorioallantoic membrane (CAM) assay as an in vivo model to study the effect of newly identified molecules on ovarian cancer invasion and metastasis. Int J Mol Sci. 2012;13(8):9959-9970. doi:10.3390/ijms13089959.

166. Shibuya M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer. 2011;2(12):1097-1105. doi:10.1177/1947601911423031.

167. Silver FH, Christiansen DL. Biomaterials Science and Biocompatability. Springer; 1999.

168. Thomson RC, Materials Research Society. Spring Meeting (1998 : San Francisco C. Biomaterials Regulating Cell Function and Tissue Development : Symposium Held April 13-14, 1998, San Francisco, California, U.S.A. Materials Research Society; 1998.

169. Douville EC, Kempczinski RF, Birinyi LK, Ramalanjaona GR. Impact of

Page 198: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

198

endothelial cell seeding on long-term patency and subendothelial proliferation in a small-caliber highly porous polytetrafluoroethylene graft. J Vasc Surg Off Publ Soc Vasc Surg [and] Int Soc Cardiovasc Surgery, North Am Chapter. 1987;5(4):544-550.

170. Dong B, Arnoult O, Smith ME, Wnek GE. Electrospinning of collagen nanofiber scaffolds from benign solvents. Macromol Rapid Commun. 2009;30(7):539-542. doi:10.1002/marc.200800634.

171. Matthews JA, Wnek GE, Simpson DG, Bowlin GL. Electrospinning of Collagen Nanofibers. Biomacromolecules. 2002;3(2):232-238. doi:10.1021/bm015533u.

172. Mazzucotelli J, Klein-Soyer C, Beretz A. Endothelial cell seeding: coating Dacron and expanded polytetrafluoroethylene vascular grafts with a biological glue allows adhesion and growth of human. J Artif …. 1991. http://europepmc.org/abstract/med/1834589. Accessed May 30, 2017.

173. Salacinski H, Tiwari A, Hamilton G. Cellular engineering of vascular bypass grafts: role of chemical coatings for enhancing endothelial cell attachment. Med Biol. 2001. http://link.springer.com/article/10.1007/BF02345431. Accessed May 30, 2017.

174. Hoffmann J, Groll J, Heuts J, et al. Blood cell and plasma protein repellent properties of star-PEG-modified surfaces. J Biomater Sci Polym Ed. 2006;17(9):985-996. doi:10.1163/156856206778366059.

175. Aoki J, Serruys PW, Van Beusekom H, et al. Endothelial progenitor cell capture by stents coated with antibody against CD34: The HEALING-FIM (Healthy Endothelial Accelerated Lining Inhibits Neointimal Growth-First in Man) registry. J Am Coll Cardiol. 2005;45(10):1574-1579. doi:10.1016/j.jacc.2005.01.048.

176. Buschmann MD, Gluzband YA, Grodzinsky AJ, Hunziker EB. Mechanical compression modulates matrix biosynthesis in chondrocyte/agarose culture. J Cell Sci. April 1995:1497-1508. http://www.ncbi.nlm.nih.gov/pubmed/7615670. Accessed April 29, 2017.

177. Glagov S, Vito R, Giddens DP, Zarins CK. Micro-architecture and composition of artery walls: relationship to location, diameter and the distribution of mechanical stress. J Hypertens Suppl. 1992;10(6):S101-S104. http://www.ncbi.nlm.nih.gov/pubmed/1432307. Accessed May 30, 2017.

178. Yang J, Webb AR, Ameer G a. Novel Citric Acid-Based Biodegradable Elastomers for Tissue Engineering. Adv Mater. 2004;16(6):511-516. doi:10.1002/adma.200306264.

179. Motlagh D, Allen JB, Hoshi R, Yang J, Lui K, Ameer G a. Hemocompatibility evaluation of poly(diol citrate) in vitro for vascular tissue engineering. J Biomed Mater Res A. 2007;81(4):771-780. doi:10.1002/jbm.a.

Page 199: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

199

180. Yang L, Fitié CFC, van der Werf KO, Bennink ML, Dijkstra PJ, Feijen J. Mechanical properties of single electrospun collagen type I fibers. Biomaterials. 2008;29(8):955-962. doi:10.1016/j.biomaterials.2007.10.058.

181. Mcclure MJ, Sell SA, Simpson DG, Bowlin GL. Electrospun Polydioxanone, Elastin, and Collagen Vascular Scaffolds: Uniaxial Cyclic Distension. 2009;4(2). http://www.jeffjournal.org/papers/Volume4/4.2Bowlin.pdf. Accessed May 29, 2017.

182. Yao Y, Liu H, Ding X, et al. Preparation and characterization of silk fibroin/poly( l -lactide-co-ε-caprolactone) nanofibrous membranes for tissue engineering applications. J Bioact Compat Polym. 2015;30(6):633-648. doi:10.1177/0883911515585185.

183. McClure MJ, Sell SA, Simpson DG, Walpoth BH, Bowlin GL. A three-layered electrospun matrix to mimic native arterial architecture using polycaprolactone, elastin, and collagen: A preliminary study. Acta Biomater. 2010;6(7):2422-2433. doi:10.1016/j.actbio.2009.12.029.

184. Boland ED. Electrospinning collagen and elastin: preliminary vascular tissue engineering. Front Biosci. 2004;9(1-3):1422. doi:10.2741/1313.

185. Eyre DR, Paz MA, Gallop PM. Cross-linking in collagen and elastin. Annu Rev Biochem. 1984;53:717-748. doi:10.1146/annurev.biochem.53.1.717.

186. Richards FM, Knowles JR. Glutaraldehyde as a protein cross-linking reagent. J Mol Biol. 1968;37(1):231-233. doi:10.1016/0022-2836(68)90086-7.

187. Cheung DT, Nimni ME. Mechanism of crosslinking of proteins by glutaraldehyde I: reaction with model compounds. Connect Tissue Res. 1982;10(2):187-199. http://www.ncbi.nlm.nih.gov/pubmed/6219858. Accessed May 30, 2017.

188. Gough JE, Scotchford CA, Downes S. Cytotoxicity of glutaraldehyde crosslinked collagen/poly(vinyl alcohol) films is by the mechanism of apoptosis. J Biomed Mater Res. 2002;61(1):121-130. doi:10.1002/jbm.10145.

189. Olde Damink LHH, Dijkstra PJ, Van Luyn MJA, Van Wachem PB, Nieuwenhuis P, Feijen J. Glutaraldehyde as a crosslinking agent for collagen-based biomaterials. J Mater Sci Mater Med. 1995;6(8):460-472. doi:10.1007/BF00123371.

190. Rüder C, Sauter T, Kratz K, et al. Influence of fibre diameter and orientation of electrospun copolyetheresterurethanes on smooth muscle and endothelial cell behaviour. Clin Hemorheol Microcirc. 2013;55(4):513-522. doi:10.3233/CH-131787.

191. Choi S-C, Choi W-K, Jung H-J, et al. Relation between hydrophilicity and cell culturing on polystyrene Petri dish modified by ion-assisted reaction. J Appl Polym Sci. 1999;73(1):41-46. doi:10.1002/(SICI)1097-4628(19990705)73:1<41::AID-

Page 200: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

200

APP5>3.0.CO;2-3.

192. Hezi-Yamit A, Sullivan C, Wong J, et al. Impact of polymer hydrophilicity on biocompatibility: Implication for DES polymer design. J Biomed Mater Res Part A. 2009;90A(1):133-141. doi:10.1002/jbm.a.32057.

193. Horbett TA, Waldburger JJ, Ratner BD, Hoffman AS. Cell adhesion to a series of hydrophili-hydrophobic copolymers studies with a spinning disc apparatus. J Biomed Mater Res. 1988;22(5):383-404. doi:10.1002/jbm.820220503.

194. Shadwick RE. Mechanical design in arteries. J Exp Biol. 1999;202(23). http://jeb.biologists.org/content/202/23/3305.short. Accessed May 30, 2017.

195. Holzapfel GA, Sommer G, Gasser CT, Regitnig P. Determination of layer-specific mechanical properties of human coronary arteries with nonatherosclerotic intimal thickening and related constitutive modeling. Am J Physiol - Hear Circ Physiol. 2005;289(5). http://ajpheart.physiology.org/content/289/5/H2048.short. Accessed May 30, 2017.

196. Teng Z, Zhang Y, Huang Y, et al. Material properties of components in human carotid atherosclerotic plaques: A uniaxial extension study. Acta Biomater. 2014;10(12):5055-5063. doi:10.1016/j.actbio.2014.09.001.

197. Karimi A, Navidbakhsh M, Shojaei A, Faghihi S. Measurement of the uniaxial mechanical properties of healthy and atherosclerotic human coronary arteries. Mater Sci Eng C. 2013;33(5):2550-2554. doi:10.1016/j.msec.2013.02.016.

198. Teng Z, Tang D, Zheng J, Woodard PK, Hoffman AH. An experimental study on the ultimate strength of the adventitia and media of human atherosclerotic carotid arteries in circumferential and axial directions. J Biomech. 2009;42(15):2535-2539. doi:10.1016/j.jbiomech.2009.07.009.

199. Hoshi RA, Van Lith R, Jen MC, Allen JB, Lapidos KA, Ameer G. The blood and vascular cell compatibility of heparin-modified ePTFE vascular grafts. Biomaterials. 2013;34(1):30-41. doi:10.1016/j.biomaterials.2012.09.046.

200. Yang J, Motlagh D, Webb AR, Ameer GA. Novel Biphasic Elastomeric Scaffold for Small-Diameter Blood. 2005;11(11):1876-1886.

201. Boland ED, Bowlin GL. Electrospinning collagen and elastin : Preliminary vascular tissue engineering. 2004;(March 2016).

202. Abbott WM, Megerman J, Hasson JE, L’Italien G, Warnock DF. Effect of compliance mismatch on vascular graft patency. J Vasc Surg. 1987;5(2):376-382. http://www.ncbi.nlm.nih.gov/pubmed/3102762. Accessed May 31, 2017.

203. Lyman DJ, Fazzio FJ, Voorhees H, Robinson G, Albo D. Compliance as a factor effecting the patency of a copolyurethane vascular graft. J Biomed Mater Res.

Page 201: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

201

1978;12(3):337-345. doi:10.1002/jbm.820120307.

204. Kinley CE, Marble AE. Compliance: a continuing problem with vascular grafts. J Cardiovasc Surg (Torino). 1980;21(2):163-170. http://www.ncbi.nlm.nih.gov/pubmed/7364859. Accessed May 31, 2017.

205. Salacinski HJ, Goldner S, Giudiceandrea A, Hamilton G, Seifalian AM. The Mechanical Behavior of Vascular Grafts: A Review. J Biomater Appl. 2001;15(3):241-278. doi:10.1106/NA5T-J57A-JTDD-FD04.

206. Niklason LE, Gao J, Abbott WM, et al. Functional arteries grown in vitro. Science. 1999;284(5413):489-493. http://www.ncbi.nlm.nih.gov/pubmed/10205057. Accessed May 31, 2017.

207. Konig G, McAllister TN, Dusserre N, et al. Mechanical properties of completely autologous human tissue engineered blood vessels compared to human saphenous vein and mammary artery. Biomaterials. 2009;30(8):1542-1550. doi:10.1016/j.biomaterials.2008.11.011.

208. Gorbet MB, Sefton M V. Biomaterial-associated thrombosis: roles of coagulation factors, complement, platelets and leukocytes. Biomaterials. 2004;25(26):5681-5703. doi:10.1016/j.biomaterials.2004.01.023.

209. Daemen J, Wenaweser P, Tsuchida K, et al. Early and late coronary stent thrombosis of sirolimus-eluting and paclitaxel-eluting stents in routine clinical practice: data from a large two-institutional cohort study. Lancet. 2007;369(9562):667-678. doi:10.1016/S0140-6736(07)60314-6.

210. Ramadugu P, Alikatte KL, Dhudipala N. A Review on Biodegradable and Bioabsorbable Stents for Coronary Artery Disease. J Bioequiv Availab. 2016;8(82):64-67. doi:10.4172/jbb.1000269.

211. Silver AE. Shear Stress-Mediated Arterial Remodeling in Atherosclerosis: Too Much of a Good Thing? Circulation. 2006;113(24):2787-2789. doi:10.1161/CIRCULATIONAHA.106.634378.

212. de Luca G, Dirksen MT, Spaulding C, Kelboek H, Schalij MJ. Drug-Eluting vs Bare-Metal Stents in Primary Angioplasty. Arch Intern Med. 2012;172(8):611-621. doi:10.1001/archinternmed.2012.758.

213. Lüscher TF, Steffel J, Eberli FR, et al. Drug-eluting stent and coronary thrombosis: Biological mechanisms and clinical implications. Circulation. 2007;115(8):1051-1058. doi:10.1161/CIRCULATIONAHA.106.675934.

214. Komori H, Tsuji T, Motohara S, et al. Humans Initial and 6-Month Results of Biodegradable Poly-l-Lactic Acid Coronary Stents in Initial and 6-Month Results of Biodegradable Poly-l-Lactic Acid Coronary Stents in Humans. Circulation. 2000;102:399-404. doi:10.1161/01.CIR.102.4.399.

Page 202: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

202

215. Uurto I, Juuti H, Parkkinen J, et al. Biodegradable Self-Expanding Poly-L/D-Lactic Acid Vascular Stent: A Pilot Study in Canine and Porcine Iliac Arteries. J Endovasc Ther. 2004;11(6):712-718. doi:10.1583/04-127MR.1.

216. Venkatraman S, Poh TL, Vinalia T, Mak KH, Boey F. Collapse pressures of biodegradable stents. Biomaterials. 2003;24(12):2105-2111. http://www.ncbi.nlm.nih.gov/pubmed/12628831. Accessed February 20, 2017.

217. VOGT F, Stein A, Rettemeier G, et al. Long-term assessment of a novel biodegradable paclitaxel-eluting coronary polylactide stent. Eur Heart J. 2004;25(15):1330-1340. doi:10.1016/j.ehj.2004.06.010.

218. Us B, Bolz A, Popp T. Implantable , bioresorbable vessel wall support , in particular coronary stent. 2001:1-9.

219. Li N, Zheng Y. Novel Magnesium Alloys Developed for Biomedical Application: A Review. J Mater Sci Technol. 2013;29(6):489-502. doi:10.1016/j.jmst.2013.02.005.

220. Diba M, Tapia F, Boccaccini AR, Strobel LA. Magnesium-Containing Bioactive Glasses for Biomedical Applications. Int J Appl Glas Sci. 2012;3(3):221-253. doi:10.1111/j.2041-1294.2012.00095.x.

221. Hornberger H, Virtanen S, Boccaccini AR. Biomedical coatings on magnesium alloys – A review. Acta Biomater. 2012;8(7):2442-2455. doi:10.1016/j.actbio.2012.04.012.

222. Berglund IS, Brar HS, Dolgova N, et al. Synthesis and characterization of Mg - Ca - Sr alloys for biodegradable orthopedic implant applications. J Biomed Mater Res Part B Appl Biomater J Biomed Mater Res Part B. 2012;100(100):1524-1534. doi:10.1002/jbm.b.32721.

223. Ida. Design and Evaluation of a Biodegradable Magnesium Alloy for use as an Implant Material. 2010.

224. Mazur A, Maier JAM, Rock E, Gueux E, Nowacki W, Rayssiguier Y. Magnesium and the inflammatory response: Potential physiopathological implications. Arch Biochem Biophys. 2007;458(1):48-56. doi:10.1016/j.abb.2006.03.031.

225. Hwang D, Yen C, Nadler J. Effect of extracellular magnesium on platelet activation and intracellular calcium mobilization. Am J. 1992. http://ajh.oxfordjournals.org/content/5/10/700.short. Accessed February 20, 2017.

226. Nadler J, Goodson S, Rude R. Evidence that prostacyclin mediates the vascular action of magnesium in humans. Hypertension. 1987. http://hyper.ahajournals.org/content/9/4/379.short. Accessed February 20, 2017.

227. Gawaz M, Ott I, Reininger A. Effects of magnesium on platelet aggregation and

Page 203: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

203

adhesion. Magnesium modulates surface expression of glycoproteins on platelets in vitro and ex vivo. Thromb. 1994. http://europepmc.org/abstract/med/7740463. Accessed February 20, 2017.

228. Ouchi Y, Tabata R, Stergiopoulos K. Effect of dietary magnesium on development of atherosclerosis in cholesterol-fed rabbits. Vasc Biol. 1990. http://atvb.ahajournals.org/content/10/5/732.short. Accessed February 20, 2017.

229. Motlagh D, Yang J, Lui KY, Webb AR, Ameer GA. Hemocompatibility evaluation of poly(glycerol-sebacate) in vitro for vascular tissue engineering. Biomaterials. 2006;27:4315-4324. doi:10.1016/j.biomaterials.2006.04.010.

230. Ramaswamy V, Dirr E, Allen J. The Effect of Simulated Microgravity on Differentiation of Porcine Blood Derived Vascular Stem Cells. Stem Cells Dev. December 2015. doi:10.1089/scd.2015.0297.

231. Ramaswamy V, Goins A, Allen JB. Altered Functions of Human Blood Derived Vascular Endothelial Cells by Simulated Microgravity. Gravitational Sp Res. 2016;4(1).

232. Stone PH, Coskun AU, Kinlay S, et al. Effect of Endothelial Shear Stress on the Progression of Coronary Artery Disease, Vascular Remodeling, and In-Stent Restenosis in Humans. Circulation. 2003;108(4).

233. Gu X, Zheng Y, Cheng Y, Zhong S, Xi T. In vitro corrosion and biocompatibility of binary magnesium alloys. Biomaterials. 2009;30(4):484-498. doi:10.1016/j.biomaterials.2008.10.021.

234. Wang MO, Etheridge JM, Thompson JA, Vorwald CE, Dean D, Fisher JP. Evaluation of the In Vitro Cytotoxicity of Cross-Linked Biomaterials. Biomacromolecules. 2013;14(5):1321-1329. doi:10.1021/bm301962f.

235. Henkelman S, Rakhorst G, Blanton J, van Oeveren W. Standardization of incubation conditions for hemolysis testing of biomaterials. Mater Sci Eng C. 2009;29(5):1650-1654. doi:10.1016/j.msec.2009.01.002.

236. Vijayanand K, Pattanayak DK, Rama Mohan TR, Banerjee R. Interpreting Blood-Biomaterial Interactions from Surface Free Energy and Work of Adhesion. Trends Biomater Artif Organs. 2005;18(2). http://www.sbaoi.org. Accessed February 21, 2017.

237. Seyfert UT, Biehl V, Schenk J. In vitro hemocompatibility testing of biomaterials according to the ISO 10993-4. Biomol Eng. 2002;19(2):91-96. doi:10.1016/S1389-0344(02)00015-1.

238. Park RS, Kim YK, Lee SJ, et al. Corrosion behavior and cytotoxicity of Mg-35Zn-3Ca alloy for surface modified biodegradable implant material. J Biomed Mater Res Part B Appl Biomater. 2012;100B(4):911-923. doi:10.1002/jbm.b.32652.

Page 204: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

204

239. Anitua E, Prado R, Orive G, Tejero R. Effects of calcium-modified titanium implant surfaces on platelet activation, clot formation, and osseointegration. J Biomed Mater Res Part A. 2015;103(3):969-980. doi:10.1002/jbm.a.35240.

240. Pors Nielsen S. The biological role of strontium. Bone. 2004;35(3):583-588. doi:10.1016/j.bone.2004.04.026.

241. Rayssiguier Y, Gueux E, Weiser D. Effect of magnesium deficiency on lipid metabolism in rats fed a high carbohydrate diet. J Nutr. 1981. http://europepmc.org/abstract/med/7299488. Accessed February 20, 2017.

242. Iseri LT, French JH. Magnesium: nature’s physiologic calcium blocker. Am Heart J. 1984;108(1):188-193. http://www.ncbi.nlm.nih.gov/pubmed/6375330. Accessed February 20, 2017.

243. Stern DM, Esposito C, Gerlach H, et al. Endothelium and regulation of coagulation. Diabetes Care. 1991;14(2):160-166. http://www.ncbi.nlm.nih.gov/pubmed/2060425. Accessed August 31, 2015.

244. Esmon CT. The interactions between inflammation and coagulation. Br J Haematol. 2005;131(4):417-430. doi:10.1111/j.1365-2141.2005.05753.x.

245. Thomas MR, Storey RF. The role of platelets in inflammation. Thromb Haemost. 2015;114(3):449-458. doi:10.1160/TH14-12-1067.

246. Piro M, Giubilato G, Pinnelli M, Giordano Sciacca P, Biasucci LM. Endothelium and inflammation. Panminerva Med. 2005;47(2):75-80.

247. Niewiarowski S, Lowery CT, Hawiger J, Millman M, Timmons S. Immunoassay of human platelet factor 4(PF4, antiheparin factor) by radial immunodiffusion. J Lab Clin Med. 1976;87(4):720-733. http://www.ncbi.nlm.nih.gov/pubmed/818325. Accessed February 20, 2017.

248. Gerotziafas GT, Elalamy I, Lecrubier C, et al. The role of platelet factor 4 in platelet aggregation induced by the antibodies implicated in heparin-induced thrombocytopenia. Blood Coagul Fibrinolysis. 2001;12(7):511-520. http://www.ncbi.nlm.nih.gov/pubmed/11685038. Accessed February 20, 2017.

249. Gutensohn K, Beythien C, Bau J, et al. In Vitro Analyses of Diamond-like Carbon Coated Stents: Reduction of Metal Ion Release, Platelet Activation, and Thrombogenicity. Thromb Res. 2000;99(6):577-585. doi:10.1016/S0049-3848(00)00295-4.

250. Klein CL, Nieder P, Wagner M, et al. The role of metal corrosion in inflammatory processes: induction of adhesion molecules by heavy metal ions. J Mater Sci Mater Med. 1994;5(11):798-807. doi:10.1007/BF00213138.

251. Colombo A, Karvouni E. Biodegradable Stents. Circulation. 2000;102(4).

Page 205: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

205

252. Lu P, Fan H, Liu Y, Cao L, Wu X, Xu X. Controllable biodegradability, drug release behavior and hemocompatibility of PTX-eluting magnesium stents. Colloids Surfaces B Biointerfaces. 2011;83(1):23-28. doi:10.1016/j.colsurfb.2010.10.038.

253. Schildberger A, Rossmanith E, Eichhorn T, Strassl K, Weber V. Monocytes, peripheral blood mononuclear cells, and THP-1 cells exhibit different cytokine expression patterns following stimulation with lipopolysaccharide. Mediators Inflamm. 2013;2013:697972. doi:10.1155/2013/697972.

254. Anderson JM, Miller KM. Biomaterial biocompatibility and the macrophage. Biomaterials. 1984;5(1):5-10. http://www.ncbi.nlm.nih.gov/pubmed/6375747. Accessed February 20, 2017.

255. Cardona MA, Simmons RL, Kaplan SS. TNF and IL-1 generation by human monocytes in response to biomaterials. J Biomed Mater Res. 1992;26(7):851-859. doi:10.1002/jbm.820260703.

256. Dinarello CA. Cytokines and Biocompatibility. Blood Purif. 2008;8(4):208-213. doi:10.1159/000169968.

257. Diya Zhang D, Lili Chen L, Shenglai Li S, Zhiyuan Gu Z, Jie Yan J. Lipopolysaccharide (LPS) of Porphyromonas gingivalis induces IL-1β, TNF-α and IL-6 production by THP-1 cells in a way different from that of Escherichia coli LPS. Innate Immun. 2008;14(2):99-107. doi:10.1177/1753425907088244.

258. Kim D-H, Lipke EA, Kim P, et al. Nanoscale cues regulate the structure and function of macroscopic cardiac tissue constructs. Proc Natl Acad Sci U S A. 2010;107(2):565-570. doi:10.1073/pnas.0906504107.

259. Feng M, Peng J, Song C, Wang Y. Mammalian cell cultivation in space. Microgravity Sci Technol. 1994;7(2):207-210. http://europepmc.org/abstract/MED/11541854. Accessed November 2, 2015.

260. Ali MH, Schumacker PT. Endothelial responses to mechanical stress: where is the mechanosensor? Crit Care Med. 2002;30(5 Suppl):S198-S206. http://www.ncbi.nlm.nih.gov/pubmed/12004236. Accessed December 25, 2015.

261. Gray ML, Pizzanelli AM, Grodzinsky AJ, Lee RC. Mechanical and physicochemical determinants of the chondrocyte biosynthetic response. J Orthop Res. 1988;6(6):777-792. doi:10.1002/jor.1100060602.

262. DiFederico E, Shelton JC, Bader DL. Complex mechanical conditioning of cell-seeded agarose constructs can influence chondrocyte biosynthetic activity. Biotechnol Bioeng. March 2017. doi:10.1002/bit.26273.

263. Carlsson SIM, Bertilaccio MTS, Ballabio E, Maier JAM. Endothelial stress by gravitational unloading: effects on cell growth and cytoskeletal organization.

Page 206: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

206

Biochim Biophys Acta. 2003;1642(3):173-179. http://www.ncbi.nlm.nih.gov/pubmed/14572900. Accessed September 3, 2015.

264. Gimbrone MA, Lelkes P. Mechanical Forces and the Endothelium. CRC Press; 2003. https://books.google.com/books?id=BCxb1JD1kRAC&pgis=1. Accessed August 31, 2015.

265. Yates B., Kerman I. Post-spaceflight orthostatic intolerance: possible relationship to microgravity-induced plasticity in the vestibular system. Brain Res Rev. 1998;28(1-2):73-82. doi:10.1016/S0165-0173(98)00028-9.

266. Convertino VA. Status of cardiovascular issues related to space flight: Implications for future research directions. Respir Physiol Neurobiol. 2009;169 Suppl :S34-S37. doi:10.1016/j.resp.2009.04.010.

267. Mukai CN, Lathers CM, Charles JB, Bennett BS. Cardiovascular responses to repetitive exposure to hyper- and hypogravity states produced by parabolic flight. J Clin Pharmacol. 1994;34(5):472-479.

268. Antonutto G, di Prampero PE. Cardiovascular deconditioning in microgravity: some possible countermeasures. Eur J Appl Physiol. 2003;90(3-4):283-291. doi:10.1007/s00421-003-0884-5.

269. Rubanyi GM. The role of endothelium in cardiovascular homeostasis and diseases. J Cardiovasc Pharmacol. 1993;22 Suppl 4:S1-S14. http://www.ncbi.nlm.nih.gov/pubmed/7523767. Accessed August 31, 2015.

270. Allen JB, Khan S, Lapidos KA, Ameer GA. Toward engineering a human neoendothelium with circulating progenitor cells. Stem Cells. 2010;28(2):318-328. doi:10.1002/stem.275.

271. Hristov M, Erl W, Weber PC. Endothelial progenitor cells: Mobilization, differentiation, and homing. Arterioscler Thromb Vasc Biol. 2003;23(7):1185-1189. doi:10.1161/01.ATV.0000073832.49290.B5.

272. Yamamoto K, Takahashi T, Asahara T, et al. Proliferation, differentiation, and tube formation by endothelial progenitor cells in response to shear stress. J Appl Physiol. 2003;95(5):2081-2088. doi:10.1152/japplphysiol.00232.2003.

273. Urbich C, Dimmeler S. Endothelial progenitor cells: Characterization and role in vascular biology. Circ Res. 2004;95(4):343-353. doi:10.1161/01.RES.0000137877.89448.78.

274. Zampetaki A, Kirton JP, Xu Q. Vascular repair by endothelial progenitor cells. Cardiovasc Res. 2008;78(3):413-421. doi:10.1093/cvr/cvn081.

275. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275(5302):964-967.

Page 207: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

207

doi:10.1126/science.275.5302.964.

276. Rotmans JI, Heyligers JMM, Stroes ESG, Pasterkamp G. Endothelial progenitor cell-seeded grafts: rash and risky. Can J Cardiol. 2006;22(13):1113-1116. doi:10.1016/S0828-282X(06)70946-1.

277. Kunz GA, Liang G, Cuculi F, et al. Circulating endothelial progenitor cells predict coronary artery disease severity. Am Heart J. 2006;152(1):190-195. doi:10.1016/j.ahj.2006.02.001.

278. Schmidt-Lucke C, Rössig L, Fichtlscherer S, et al. Reduced Number of Circulating Endothelial Progenitor Cells Predicts Future Cardiovascular Events. Circulation. 2005;111(22). http://circ.ahajournals.org/content/111/22/2981.short. Accessed April 29, 2017.

279. Murasawa S, Asahara T. Endothelial progenitor cells for vasculogenesis. Physiology (Bethesda). 2005;20:36-42. doi:10.1152/physiol.00033.2004.

280. Kalka C, Masuda H, Takahashi T, et al. Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc Natl Acad Sci U S A. 2000;97(7):3422-3427. doi:10.1073/pnas.97.7.3422.

281. Möbius-Winkler S, Höllriegel R, Schuler G, Adams V. Endothelial progenitor cells: Implications for cardiovascular disease. Cytom Part A. 2009;75A(1):25-37. doi:10.1002/cyto.a.20669.

282. Radtke AL, Herbst-Kralovetz MM. Culturing and applications of rotating wall vessel bioreactor derived 3D epithelial cell models. J Vis Exp. 2012;(62). doi:10.3791/3868.

283. Lelovas PP, Kostomitsopoulos NG, Xanthos TT. A comparative anatomic and physiologic overview of the porcine heart. J Am Assoc Lab Anim Sci. 2014;53(5):432-438. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4181683&tool=pmcentrez&rendertype=abstract. Accessed August 31, 2015.

284. Morbidelli L, Monici M, Marziliano N, et al. Simulated hypogravity impairs the angiogenic response of endothelium by up-regulating apoptotic signals. Biochem Biophys Res Commun. 2005;334(2):491-499. doi:10.1016/j.bbrc.2005.06.124.

285. Versari S, Villa A, Bradamante S, Maier JAM. Alterations of the actin cytoskeleton and increased nitric oxide synthesis are common features in human primary endothelial cell response to changes in gravity. Biochim Biophys Acta. 2007;1773(11):1645-1652. doi:10.1016/j.bbamcr.2007.05.014.

286. Carlsson SIM, Bertilaccio MTS, Ascari I, Bradamante S, Maier JAM. Modulation of human endothelial cell behaviour in simulated microgravity. J Gravit Physiol. 2002;9(1):P273-P274. http://www.ncbi.nlm.nih.gov/pubmed/15002577. Accessed

Page 208: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

208

September 3, 2015.

287. Liang C-C, Park AY, Guan J-L. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2007;2(2):329-333. doi:10.1038/nprot.2007.30.

288. Cogoli A. The effect of hypogravity and hypergravity on cells of the immune system. J Leukoc Biol. 1993;54(3):259-268.

289. Zhang Y, Sang C, Paulsen K, et al. ICAM-1 expression and organization in human endothelial cells is sensitive to gravity. Acta Astronaut. 2010;67(9-10):1073-1080. doi:10.1016/j.actaastro.2010.06.027.

290. Uva B, Masini M, Sturla M, Prato P. Clinorotation-induced weightlessness influences the cytoskeleton of glial cells in culture. Brain Res. 2002;934(2):132-139. http://www.sciencedirect.com/science/article/pii/S0006899302024150. Accessed August 31, 2015.

291. Ritz BW, Lelkes PI, Gardner EM. Functional recovery of peripheral blood mononuclear cells in modeled microgravity. FASEB J. 2006;20(2):305-307. doi:10.1096/fj.04-3122fje.

292. MacEneaney OJ, Kushner EJ, Van Guilder GP, Greiner JJ, Stauffer BL, DeSouza CA. Endothelial progenitor cell number and colony-forming capacity in overweight and obese adults. Int J Obes (Lond). 2009;33(2):219-225. doi:10.1038/ijo.2008.262.

293. Zhang H, Vakil V, Braunstein M, et al. Circulating endothelial progenitor cells in multiple myeloma: implications and significance. Blood. 2005;105(8):3286-3294. doi:10.1182/blood-2004-06-2101.

294. Plett PA, Abonour R, Frankovitz SM, Orschell CM. Impact of modeled microgravity on migration, differentiation, and cell cycle control of primitive human hematopoietic progenitor cells. Exp Hematol. 2004;32(8):773-781. doi:10.1016/j.exphem.2004.03.014.

295. Fuchs B, Medvedev A. Countermeasures for ameliorating in-flight immune dysfunction. J Leukoc Biol. 1993;54(3):245-252. http://www.jleukbio.org/content/54/3/245.abstract. Accessed August 31, 2015.

296. Sonnenfeld G. The immune system in space, including Earth-based benefits of space-based research. Curr Pharm Biotechnol. 2005;6(4):343-349. http://www.ncbi.nlm.nih.gov/pubmed/16101473. Accessed November 4, 2015.

297. Melchiorri AJ, Hibino N, Fisher JP. Strategies and techniques to enhance the in situ endothelialization of small-diameter biodegradable polymeric vascular grafts. Tissue Eng Part B Rev. 2013;19(4):292-307. doi:10.1089/ten.TEB.2012.0577.

Page 209: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

209

298. Gerritsen ME. Functional heterogeneity of vascular endothelial cells. Biochem Pharmacol. 1987;36(17):2701-2711. http://www.ncbi.nlm.nih.gov/pubmed/2820420. Accessed August 31, 2015.

299. van Hinsbergh VWM. Endothelium--role in regulation of coagulation and inflammation. Semin Immunopathol. 2012;34(1):93-106. doi:10.1007/s00281-011-0285-5.

300. Grenon SM, Jeanne M, Aguado-Zuniga J, Conte MS, Hughes-Fulford M. Effects of gravitational mechanical unloading in endothelial cells: association between caveolins, inflammation and adhesion molecules. Sci Rep. 2013;3:1494. doi:10.1038/srep01494.

301. Carlsson SIM, Bertilaccio MTS, Ascari I, Bradamante S, Maier JAM. Modulation of human endothelial cell behaviour in simulated microgravity. J Gravit Physiol. 2002;9(1):P273-P274. http://europepmc.org/abstract/med/15002577. Accessed November 11, 2015.

302. Cotrupi S, Maier JAM. Is HSP70 upregulation crucial for cellular proliferative response in simulated microgravity? J Gravit Physiol. 2004;11(2):P173-P176. http://europepmc.org/abstract/med/16237827. Accessed December 11, 2015.

303. Kiang J. Heat Shock Protein 70 kDa Molecular Biology, Biochemistry, and Physiology. Pharmacol Ther. 1998;80(2):183-201. doi:10.1016/S0163-7258(98)00028-X.

304. Beere H. Stress management – heat shock protein-70 and the regulation of apoptosis. Trends Cell Biol. 2001;11(1):6-10. doi:10.1016/S0962-8924(00)01874-2.

305. Ravagnan L, Gurbuxani S, Susin SA, et al. Heat-shock protein 70 antagonizes apoptosis-inducing factor. Nat Cell Biol. 2001;3(9):839-843. doi:10.1038/ncb0901-839.

306. Gamboa A, Okamoto LE, Raj SR, et al. Nitric oxide and regulation of heart rate in patients with postural tachycardia syndrome and healthy subjects. Hypertension. 2013;61(2):376-381. doi:10.1161/HYPERTENSIONAHA.111.00203.

307. Spisni E, Bianco MC, Griffoni C, et al. Mechanosensing role of caveolae and caveolar constituents in human endothelial cells. J Cell Physiol. 2003;197(2):198-204. doi:10.1002/jcp.10344.

308. Vaziri ND, Ding Y, Sangha DS, Purdy RE. Upregulation of NOS by simulated microgravity, potential cause of orthostatic intolerance. J Appl Physiol. 2000;89(1):338-344. http://www.ncbi.nlm.nih.gov/pubmed/10904069. Accessed December 11, 2015.

309. Trepels T, Zeiher AM, Fichtlscherer S. The endothelium and inflammation.

Page 210: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

210

Endothelium. 2009;13(6):423-429. doi:10.1080/10623320601061862.

310. Boonyaratanakornkit JB, Cogoli A, Li C-F, et al. Key gravity-sensitive signaling pathways drive T cell activation. FASEB J. 2005;19(14):2020-2022. doi:10.1096/fj.05-3778fje.

311. Hughes-Fulford M. To infinity ... and beyond! Human spaceflight and life science. FASEB J. 2011;25(9):2858-2864. doi:10.1096/fj.11-0902ufm.

312. May LT, Torcia G, Cozzolino F, et al. Interleukin-6 gene expression in human endothelial cells: RNA start sites, multiple IL-6 proteins and inhibition of proliferation. Biochem Biophys Res Commun. 1989;159(3):991-998. doi:10.1016/0006-291X(89)92206-7.

313. Deakin AM, Payne AN, Whittle BJ, Moncada S. The modulation of IL-6 and TNF-alpha release by nitric oxide following stimulation of J774 cells with LPS and IFN-gamma. Cytokine. 1995;7(5):408-416. doi:10.1006/cyto.1995.0056.

314. Orshal JM, Khalil RA. Reduced endothelial NO-cGMP-mediated vascular relaxation and hypertension in IL-6-infused pregnant rats. Hypertension. 2004;43(2):434-444. doi:10.1161/01.HYP.0000113044.46326.98.

315. Borges TJ, Wieten L, van Herwijnen MJC, et al. The anti-inflammatory mechanisms of Hsp70. Front Immunol. 2012;3:95. doi:10.3389/fimmu.2012.00095.

316. Jacquier-Sarlin MR, Fuller K, Dinh-Xuan AT, Richard M-J, Polla BS. Protective effects of hsp70 in inflammation. Experientia. 1994;50(11-12):1031-1038. doi:10.1007/BF01923458.

317. Luo X, Zuo X, Zhou Y, et al. Extracellular heat shock protein 70 inhibits tumour necrosis factor-alpha induced proinflammatory mediator production in fibroblast-like synoviocytes. Arthritis Res Ther. 2008;10(2):R41. doi:10.1186/ar2399.

318. Van Guilder GP, Hoetzer GL, Smith DT, et al. Endothelial t-PA release is impaired in overweight and obese adults but can be improved with regular aerobic exercise. Am J Physiol Endocrinol Metab. 2005;289(5):E807-E813. doi:10.1152/ajpendo.00072.2005.

319. Levin EG, Marzec U, Anderson J, Harker LA. Thrombin stimulates tissue plasminogen activator release from cultured human endothelial cells. J Clin Invest. 1984;74(6):1988-1995. doi:10.1172/JCI111620.

320. Maiello M, Boeri D, Podesta F, et al. Increased Expression of Tissue Plasminogen Activator and Its Inhibitor and Reduced Fibrinolytic Potential of Human Endothelial Cells Cultured in Elevated Glucose. Diabetes. 1992;41(8):1009-1015. doi:10.2337/diab.41.8.1009.

Page 211: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

211

321. Nickerson CA, Pellis NR (Neal R, Ott CM. Effect of Spaceflight and Spaceflight Analogue Culture on Human and Microbial Cells : Novel Insights into Disease Mechanisms.

322. Mazzolai Karima Bouzourene Daniel Hayoz Françoise Dignat-George Jia Wei Liu Henri Bounameaux Sylvie Dunoyer-Geindre Egbert Kruithof LK. Characterization of Human Late Outgrowth Endothelial Progenitor-Derived Cells under Various Flow Conditions. J Vasc Res. 2011;48:443-451. doi:10.1159/000324844.

323. Patrick CW, Mikos AG, McIntire L V. Frontiers in Tissue Engineering. Pergamon; 1998.

324. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13(1):9-22. http://www.ncbi.nlm.nih.gov/pubmed/9872925.

325. Klagsbrun M, D’Amore PA. Vascular endothelial growth factor and its receptors. Cytokine Growth Factor Rev. 1996;7(3):259-270. doi:S1359610196000275 [pii].

326. Sethi R, Lee CH. Endothelial progenitor cell capture stent: Safety and effectiveness. J Interv Cardiol. 2012;25(5):493-500. doi:10.1111/j.1540-8183.2012.00740.x.

327. Nipun Babu V, Kannan S. Enhanced delivery of baicalein using cinnamaldehyde cross-linked chitosan nanoparticle inducing apoptosis. Int J Biol Macromol. 2012;51(5):1103-1108. doi:10.1016/j.ijbiomac.2012.08.038.

328. Cheirmadurai K, Thanikaivelan P, Murali R. Highly biocompatible collagen–Delonix regia seed polysaccharide hybrid scaffolds for antimicrobial wound dressing. Carbohydr Polym. 2016;137:584-593. doi:10.1016/j.carbpol.2015.11.015.

Page 212: © 2017 Vidhya Ramaswamy5 with great appreciation my brother, Dr. Bharath Ramaswamy, who has been, a great source of inspiration, a dear friend, philosopher and a wise guide. Without

212

BIOGRAPHICAL SKETCH

Vidhya was born in the beautiful metropolitan city of Chennai, home to the

world’s longest natural urban beach. She spent the major part of her life growing up in

this culturally rich city with her family. She graduated high school from Padma Seshadri

Bala Bhavan and then moved to Tiruchirapalli in south India for her bachelor’s degree.

Her alma mater was the National Institute of Technology where she earned a Bachelor

of Technology degree in Metallurgical and Materials Engineering in 2013. She then

moved to Florida to attend graduate school at University of Florida in fall 2013. She

started as a master’s student and transitioned to the doctoral program under the

guidance of Dr. Josephine Allen in fall 2014. She received her Master of Science

degree in 2016 and her doctoral degree in 2017, in materials Science and engineering

with a concentration in biomaterials. She has also served as the secretary for Society of

Biomaterials UF chapter during 2016-2017 and as an executive member of the student

safety council at the Department of Materials Science and Engineering.