Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of...

129
Characterization of Changes in Extracellular Matrix and Cellular Phenotypes in Early Calcific Aortic Valve Disease by Andrea Victoria Kwong A thesis submitted in conformity with the requirements for the degree of Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Andrea Victoria Kwong 2014

Transcript of Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of...

Page 1: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

Characterization of Changes in Extracellular Matrix and Cellular Phenotypes in Early Calcific Aortic Valve Disease

by

Andrea Victoria Kwong

A thesis submitted in conformity with the requirements for the degree of Master of Applied Science in Biomedical Engineering

Institute of Biomaterials and Biomedical Engineering University of Toronto

© Copyright by Andrea Victoria Kwong 2014

Page 2: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

ii

Characterization of Changes in Extracellular Matrix and Cellular

Phenotypes in Early Calcific Aortic Valve Disease

Andrea Victoria Kwong

Master of Applied Science in Biomedical Engineering

Institute of Biomaterials and Biomedical Engineering

University of Toronto

2014

Abstract

Calcific aortic valve disease (CAVD) is a prevalent disease associated with severe clinical

outcomes and without an effective medical treatment. While advanced disease is well-

characterized, there is an unmet scientific need to understand the active pathobiological

processes that promote eventual calcification and stenosis of the valve. Using a porcine model of

early CAVD, histological staining was used to identify the changes that occur in the extracellular

matrix (ECM) with a focus on proteoglycan content. Putatively more advanced lesions were

morphologically dense with increased biglycan, decreased hyaluronan, and higher ApoB score

and Sox9 fraction. These microenvironmental changes were used to delineate populations of

valve interstitial cells (VICs) for microarray analysis. Differentially upregulated genes from the

top of lesions and non-lesion fibrosa were related to lipid accumulation, inflammatory response,

osteochondrogenesis, and ECM remodeling. An improved understanding of early CAVD

microenvironment and VIC phenotypes lays the foundation to identify novel therapeutic targets.

Page 3: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

iii

Acknowledgments

First and foremost, I would like to thank Dr. Craig Simmons for giving me this opportunity and

providing his guidance and support through the past few years. I feel extremely lucky to have

had such an approachable, patient, and enthusiastic supervisor, who is an all-around good guy. I

cannot imagine completing this thesis without Craig’s helpful insights and words of

encouragement, which always managed to breathe new life into my experiments when seemingly

nothing was working. I thoroughly enjoyed completing this project under Craig’s supervision.

I am also grateful to have been part of the Cellular Mechanobiology lab. Specifically, I would

like to thank Krista for showing me the ropes (and the joys of histology) from my first day in the

lab, Mark for being cynical with me and showing me how to do valve and RNA isolations, and

Zahra for being the best lab mom, helping me with just about everything in the lab. I would also

like to thank everyone else in the lab, past and present, for making it such a fun and positive

environment to come to every day. I have created fond memories over the last three years and

will miss all the laughs and shenanigans.

Outside of the lab, there are a number of people who provided their technical expertise to help

me complete my project. I would like to thank Brent Steer from the Marsden lab for teaching me

how to use the cryostat and laser capture microdissection system and Patrick Yau, Carl Virtanen,

Gurbaksh Basi, and Natalie Stickle from the Ontario Cancer Institute Genomics Centre for

helping me set up, process, and analyze the microarrays. I also very much appreciate the support

and insight of my committee members: Dr. Michelle Bendeck, Dr. Rita Kandel, and Dr. Eli

Sone.

Last, but not least, I would like to thank my friends and family for their support and

encouragement through these tough but satisfying years. We did it!

Page 4: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

iv

Table of Contents

Acknowledgements ....................................................................................................................... iii

Table of Contents .......................................................................................................................... iv

List of Tables ............................................................................................................................... vii

List of Figures ............................................................................................................................. viii

Chapter 1 – Thesis Motivation and Overview ............................................................................... 1

1.1 Motivation ..................................................................................................................... 1

1.2 Thesis Overview ............................................................................................................ 3

Chapter 2 – Literature Review ....................................................................................................... 4

2.1 Normal Aortic Valve Structure and Function ............................................................... 4

2.1.1 Function and Macrostructure of the Healthy Aortic Valve ........................................ 4

2.1.2 Microstructure of the Health Aortic Valve ................................................................. 4

2.1.2.1 Extracellular matrix components ................................................................... 5

2.1.2.2 Cellular components ...................................................................................... 7

2.2 Calcific Aortic Valve Disease ....................................................................................... 7

2.2.1 Histopathology of Human CAVD .............................................................................. 8

2.2.1.1 Early CAVD lesions ....................................................................................... 8

2.2.1.2 Advanced CAVD lesions ............................................................................... 9

2.2.1.2.1 Lipid accumulation .......................................................................... 10

2.2.1.2.2 Inflammatory processes ................................................................... 10

Page 5: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

v

2.2.1.2.3 Extracellular matrix remodeling ...................................................... 11

2.2.1.2.4 Calcification ..................................................................................... 13

2.2.2 Osteochondrogenic VIC changes ............................................................................. 13

2.2.3 Proteoglycans and glycosaminoglycans in CAVD ................................................... 15

2.2.3.1 Implications of the role of PGs/GAGs from atherosclerosis ....................... 16

2.2.3.2 Localization and function of specific PGs/GAGs in CAVD ....................... 18

2.2.4 Porcine models of calcific aortic valve disease ........................................................ 21

2.2.4.1 A porcine model of early calcific aortic valve disease ................................ 22

Chapter 3 – Hypotheses and Objectives ...................................................................................... 24

Chapter 4 – Proteoglycan and glycosaminoglycan content in lesions of early CAVD ............... 25

4.1 Introduction ................................................................................................................. 25

4.2 Materials and Methods ................................................................................................ 26

4.2.1 Porcine model of early calcific aortic valve disease ....................................... 26

4.2.2 Porcine leaflet handling .................................................................................. 27

4.2.3 Histological and immunohistochemical staining ............................................ 27

4.2.4 Data retrieval and statistical analyses ............................................................. 29

4.3 Results ......................................................................................................................... 32

4.3.1 HF/HC diet alters lesion ECM morphology with temporal differences ......... 32

4.3.2 Lesions that are ApoB-positive and contain Sox9-expressing cells display

unique PG/GAG composition .......................................................................... 34

Page 6: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

vi

4.3.3 Lesions with dense morphology display more advanced characteristics of

CAVD .............................................................................................................. 37

4.4 Discussion .................................................................................................................... 39

4.4.1 Biglycan may be involved in lipid retention and chondrogenesis in early

lesions of CAVD ............................................................................................. 39

4.4.2 Hyaluronan may play a protective role in early lesion pathogenesis .............. 40

4.4.3 Early CAVD lesions demonstrate further ECM remodeling with distinct

morphological characteristics .......................................................................... 41

4.5 Conclusion ................................................................................................................... 43

Chapter 5 – Phenotypes of valve interstitial cells in lesions of early CAVD .............................. 44

5.1 Introduction ................................................................................................................. 44

5.2 Materials and Methods ................................................................................................ 45

5.2.1 Frozen valve leaflet section preparation ......................................................... 45

5.2.2 Histological and immunohistochemical identification of lesions and samples of

interest ............................................................................................................. 46

5.2.3 Laser capture microdissection ......................................................................... 47

5.2.4 RNA isolation, amplification, and microarray analysis .................................. 48

5.2.5 Data processing and statistical analyses .......................................................... 49

5.2.6 Venn diagram analysis .................................................................................... 50

5.3 Results ......................................................................................................................... 50

5.3.1 Sample characterization .................................................................................. 50

5.3.2 Lesion and non-lesion VIC differential gene expression ................................ 52

Page 7: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

vii

5.3.3 Differential gene expression of VICs within lesion areas ............................... 53

5.3.4 Differential gene expression of VICs from specific lesion areas and non-lesion

areas .......................................................................................................................... 56

5.4 Discussion .................................................................................................................... 58

5.5 Conclusion ................................................................................................................... 61

Chapter 6 – Conclusions and Future Work .................................................................................. 62

6.1 Conclusions ................................................................................................................. 62

6.2 Future Work ................................................................................................................. 63

6.2.1 Further characterization of ECM changes in early CAVD lesions ................. 63

6.2.2 Validation and pathway analysis of microarray results .................................. 64

6.2.3 In vitro studies of biglycan influence on VIC function .................................. 64

6.2.4 Mechanistic studies of hyaluronan interaction with VICs ................................ 65

References .................................................................................................................................... 66

Appendix A. Supplemental Data ................................................................................................. 84

A.1 Myofibroblast detection in porcine valve lesions ....................................................... 84

A.2 Porcine model diet formulation .................................................................................. 86

A.3 PG/GAG scoring validation and analyses .................................................................. 88

A.4 Specific PG/GAG-rich lesions display distinct morphological characteristics .......... 89

A.5 RNA and cDNA quality control before microarray analysis ..................................... 90

A.6 Quality control plots for microarray analyses ............................................................ 94

A.7 Hierarchical clustering of repeated-measures ANOVA results .................................. 96

Page 8: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

viii

Appendix B. Protocols ................................................................................................................. 98

B.1 Valve leaflet histological processing for paraffin-embedded leaflets ........................ 98

B.2 Movat’s pentachrome staining for formalin-fixed, paraffin-embedded sections ...... 100

B.3 (Immuno)histochemistry for formalin-fixed, paraffin-embedded sections .............. 102

B.4 Image processing ...................................................................................................... 106

B.5 Valve leaflet histological processing for OCT-embedded leaflets ........................... 109

B.6 Movat’s pentachrome staining for frozen OCT-embedded sections ........................ 110

B.7. Immunohistochemistry staining for frozen OCT-embedded sections ..................... 112

B.8. Laser capture microdissection protocol ................................................................... 114

B.9. RNA isolation protocol ............................................................................................ 118

Page 9: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

ix

List of Tables

Table 2.1. Transcription factors upregulated in CAVD and their roles in chondrogenic and/or

osteogenic processes .................................................................................................................... 15

Table 5.1. Expression levels of select macrophage-specific markers .......................................... 51

Table 5.2. Select differentially expressed genes between lesion (top and bottom) and non-lesion

areas ............................................................................................................................................. 53

Table 5.3. Select lipid-related genes that are upregulated in the top of lesions ........................... 54

Table 5.4. Select immune-related genes that are upregulated in the top of lesions ..................... 55

Table 5.5. Select ECM remodeling-related genes that are upregulated in the top of lesions ...... 56

Table 5.6. Select differentially expressed genes that are upregulated in the top of lesions

compared to non-lesion areas ....................................................................................................... 57

Page 10: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

x

List of Figures

Figure 2.1. Healthy aortic valves are composed of a heterogeneous tri-layered structure ............ 6

Figure 2.2. PG/GAG staining intensities within and surrounding calcified nodules ................................ 12

Figure 4.1. Qualitative classification of lesion morphology ........................................................ 29

Figure 4.2. Semi-quantative scoring system for ApoB and PG/GAG content ............................ 31

Figure 4.3. HF/HC diet alters lesion morphology with temporal differences ............................. 33

Figure 4.4. Temporal changes in lesion ECM composition ......................................................... 34

Figure 4.5. Relationship between ApoB and PG/GAG score in lesion areas .............................. 35

Figure 4.6. PG-rich lesions are associated with putative chondrogenesis ................................... 36

Figure 4.7. Relationship between Sox9 fraction and PG/GAG score in lesion areas .................. 37

Figure 4.8. Dense and diffuse lesion characteristics .................................................................... 38

Figure 5.1. Cryosectioning slide schematic for each porcine sample .......................................... 46

Figure 5.2. Lesions of interest for differential gene expression analysis ..................................... 48

Figure 5.3. Distribution of differentially expressed transcripts in lesion and non-lesion areas ... 52

Figure 5.4. Transcript expression in lesion areas ......................................................................... 52

Page 11: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

1

Chapter 1

1 Thesis Overview

1.1 Motivation

Calcific aortic valve disease (CAVD) is the most common valve disease in North America and

Europe [1]. In the United States, valvular heart diseases account for over 22,000 deaths per year

with CAVD comprising almost 15,000 of those deaths annually [2]. CAVD covers a wide

spectrum of pathological changes, including aortic valve sclerosis and the more advanced form

of aortic stenosis. In 1997, the Cardiovascular Health Study (CHS) reported that early sclerosis

affects 26% of the population over 65 years of age, while late stenosis is present in 2% of this

same population [3]. The incidence almost doubles for individuals over the age of 85, in which

sclerosis is present in 48% and stenosis in 4% [3]. In a follow-up study with the same

participants, the percentage of individuals with stenosis remained the same, but those with

sclerosis increased to 29%, indicating an increase in the prevalence of CAVD [4, 5]. Along with

an increasing prevalence, CAVD is known to be associated with several poor clinical

consequences [4]. Progression from sclerosis to stenosis affects 9% of elderly patients [5] and is

associated with an 80% five-year risk of progression to heart failure, valve replacement, or death

[4].

With a high prevalence and negative clinical outcomes, there are still no effective medical

treatments for CAVD. Currently, the only medical intervention is surgical replacement of the

valve, which has its disadvantages due to the invasiveness of surgery and problems with regards

to durability and lifelong anti-coagulant administration with biological and mechanical valves,

respectively [6]. Although the cholesterol-lowering statins have been explored as a potential

therapy because of their success in treating atherosclerosis, randomized controlled studies have

yet to show their effectiveness in the treatment of valve disease [7-9]. An improved

understanding of CAVD pathogenesis is essential to satisfy the unmet need for effective medical

treatments.

Once thought to be a disease caused by general “wear-and-tear”, CAVD progression is now

recognized as an active process hallmarked by changes in the organization, composition and

Page 12: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

2

mechanical properties of the valve extracellular matrix (ECM) [10]. This results in thickening

and stiffening of the leaflet, ultimately leading to obstruction of blood flow and impaired cardiac

function. Specifically, lesions and calcification predominantly occur on the aortic side of leaflets

[10, 11]. This side-specific pathosusceptibility is not fully understood, but may give insight into

the regulatory mechanisms involved in CAVD progression. Advanced valve disease has been

studied extensively, and is well-characterized by ECM disorganization, phenotypic changes in

valvular interstitial cells (VICs), lipid infiltration, inflammation, and calcification [12, 13]. In

contrast, early pathological alterations in the cellular and ECM composition of the aortic valve

and their role in disease initiation and progression are poorly understood. As such, the study of

early CAVD pathogenesis will provide a more complete picture of whole disease progression.

A feature of CAVD is aberrant regional expression of proteoglycans (PGs) and

glycosaminoglycans (GAGs), suggesting roles for PGs/GAGs in CAVD pathogenesis. In normal

valve tissue, PGs/GAGs are a major component of only the middle spongiosa layer, but in

advanced disease, increases in the PGs, biglycan, decorin, and versican, as well as the non-

sulfated GAG hyaluronan, have been observed surrounding calcified nodules in the fibrosa layer

[14]. Overall, the role of PGs and GAGs in CAVD has not been extensively studied. It is

suspected they may aid in the initiation of disease by retaining lipids and binding macrophages

as they do in atherosclerosis [15].

PG-rich lesions similar to those seen in the fibrosa layer of human diseased aortic valves [16, 17]

have been recapitulated recently in a porcine model of early aortic valve disease by Sider et al

[18, 19]. These lesions were observed before the appearance of lipid deposition, myofibroblasts,

certain inflammatory cells, and osteoblasts, suggesting that PG lesion formation is an initial step

that occurs before inflammation and VIC activation. Moreover, these early lesions were softer

than the collagen-rich healthy fibrosa, and PG content was positively correlated with expression

of Sox9, a chondrogenic transcription factor. Structural and compositional changes in the valve

ECM can alter VIC fate and function [20, 21] and therefore, soft PG-rich matrices may be

permissive to chondrogenic VIC differentiation. PGs may also indirectly alter VIC phenotypes

by mediating lipoprotein retention and the production of oxidized lipid byproducts that induce

inflammation and calcification. Using our porcine model of early aortic valve disease, I

addressed these issues in my thesis in two complementary studies that (1) examined the

localization of specific PGs/GAGs within early lesion areas using immunohistochemistry; and

Page 13: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

3

(2) examined VIC phenotypes in altered lesion environments by microarray analysis. These

studies provide new insights into the characteristics and pathobiological processes in early

CAVD.

1.2 Thesis Overview

This thesis is organized into six chapters. Chapter 1 provides the motivation for and overview of

the thesis. Chapter 2 presents a literature review of the topics relevant to this thesis, including

basic aortic valve structure and function and the current understanding of CAVD with a focus on

histopathology, osteochondrogenic processes, and the potential role of proteoglycans in early

disease progression. Chapter 3 states the hypotheses and objectives of this study. Chapter 4

describes the temporal changes of specific proteoglycans and hyaluronan, as well as the

relationship of these extracellular matrix components with markers of lipid retention and putative

chondrogenesis. Chapter 5 describes the lipid-, immune-, osteochondrogenic-, and ECM

remodeling-related phenotypic changes in valve interstitial cells within these lesion areas.

Chapter 6 summarizes the results and provides recommendations for future work.

Page 14: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

4

Chapter 2

2 Literature Review

2.1 Normal Aortic Valve Function and Structure

2.1.1 Function and Macrostructure of the Healthy Aortic Valve

Located between the left ventricle and the aorta, the aortic valve (AV) is responsible for

preventing backflow of oxygenated blood as it leaves the heart and enters the systemic

circulation. The AV is typically made up of three semilunar cusps or leaflets: the right coronary,

the left coronary, and the noncoronary, which are named according to their relationship to the

coronary ostia. Along the top of each leaflet is the free edge or lannula. Each leaflet attaches to

the aortic wall in a crescent-shaped manner starting from the ends of the free edge, known as the

commissures, and along the basal attachment. Behind each leaflet on the outflow side are dilated

indentations in the aortic root known as the sinuses of Valsalva, which are important for creating

the necessary conditions for valve closure. The non-coapting middle region of the valve leaflet is

known as the belly.

The AV opens and closes approximately 40 million times a year and 3 billion times within an

average lifetime [22]. Valve mechanics largely depend on the changes in hemodynamic forces

and pressure gradients that occur throughout the cardiac cycle. When the left ventricle contracts

during systole, high pressure accelerates oxygenated blood outward from the heart and pushes

the leaflets open. As the left ventricle relaxes in diastole, vortices form in the sinuses of

Valsalva, which stretch the leaflets and cause them to seal along the line of coaptation, allowing

the left ventricle to fill. Apposition of the fibrous nodule of Arantius in the middle of each free

edge ensures complete closure of the valve during diastole. Failure of these mechanisms can lead

to often serious health complications.

2.1.2 Microstructure of the Healthy Aortic Valve

Healthy human aortic valve leaflets are normally composed of three distinct layers: the aortic-

side fibrosa, the middle spongiosa, and the ventricular-side ventricularis (Figure 2.1). The layers

of the valve are heterogeneous: the fibrosa layer is primarily composed of collagen, the

spongiosa is mostly PGs and GAGs, and the ventricularis contains elastin fibers in addition to

Page 15: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

5

collagen [22, 23]. The two major cell components residing in the valve are valvular endothelial

cells (VECs) and valvular interstitial cells (VICs).

2.1.2.1 Extracellular matrix components

Proper valve function is highly dependent on the complex microstructure of the ECM. Normal

valves are predominantly composed of type I collagen and significant amounts of type III

collagen. Collagen type I is found mainly in the fibrosa layer oriented circumferentially, while

collagen type III is ubiquitously spread throughout the three layers in a less organized fashion

[24, 25]. Collagen fibers give mechanical strength to the valve, allowing expansion during valve

closure in diastole and providing strength to the ventricularis backbone by transferring load to

the aortic root wall [24-26].

Elastin is mostly found in the ventricularis layer, typically found in sheets that stretch radially

from the base of the leaflet to the line of coaptation [25, 27]. Overall, elastin fibers provide

flexibility to the leaflets, allowing repeated deformation and reformation as the valve opens and

closes. In the fibrosa layer, elastin forms honeycomb-like structures, suggesting that they may be

linked and mechanically coupled to collagen fibers. It is believed that the primary role of elastin

fibers involves maintaining valve architecture by returning collagen fibers to their resting

crimped state between loading cycles [24, 27, 28].

Proteoglycans are another critical ECM component and consist of a core protein covalently

linked to at least one GAG chain. GAG chains are repeating disaccharide units that contribute a

negative charge to the PG core and consequently, are an important contributor to PG function

[29-31]. The major types of PGs/GAGs in the valve are biglycan, decorin, versican, and

hyaluronan. Biglycan and decorin are both small, leucine-rich PGs and consist of chondroitin

sulfate and/or dermatan sulfate GAG chains, while versican is a large chondroitin sulfate PG.

Hyaluronan is a type of GAG and is unique because it is the only GAG that exists freely in the

body, separate from a PG chain. Total GAG composition in the valve is approximately 55%

hyaluronan, 30% chondroitin-4-sulfate/chondroitin-6-sulfate, and 15% dermatan sulfate [32, 33].

Although PGs/GAGs are predominantly found in the spongiosa layer, they are also present in the

other valve layers. Histological studies demonstrate unique PG/GAG localization patterns in

each of the valve layers. Decorin and biglycan are ubiquitously found throughout the valve

Page 16: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

6

layers, but most strongly expressed in the elastin-rich ventricularis layer, suggesting they may

play a role in maintaining leaflet tension and elastogenesis [26, 28]. These small, leucine-rich

PGs also frequently co-localize with collagen, suggesting a role in collagen fibrillogenesis [14,

34]. The large chondroitin sulfate PG versican and non-sulfated GAG hyaluronan are most

abundant in the spongiosa, where they are thought to provide resistance to cyclic compression

and provide lubrication to the outer fibrosa and ventricularis layers by keeping the spongiosa

properly hydrated [14, 26]. Overall, it is believed PGs are necessary for stable assembly of the

ECM and functional cell-ECM interactions.

Figure 2.1. Healthy aortic valves are composed of a heterogeneous tri-layered structure. Movat’s

pentachrome staining (blue = proteoglycan, yellow = collagen, red = cytoplasm, black = elastin/nuclei) of

a porcine aortic valve that is representative of the tri-layered structure in humans. Scale bar = 100 µm.

The valvular ECM architecture is not only critical in maintaining the functional mechanics of the

cardiac cycle, but is also a crucial component that transduces micromechanical forces into

molecular changes that mediate normal valve cell function and biology. Valve dysfunction

occurs when there is disruption of the tri-layered valve structure, resulting from maladaptive

ECM protein regulation, changes in quantity and distribution of ECM components, and

expression of ECM components usually only involved in development or osteochondrogenesis

Fibrosa

Spongiosa

Ventricularis

Page 17: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

7

[35]. Such environmental changes likely affect valve cell phenotype and differentiation, which

are known to be affected by both biomechanical and microenvironmental cues [36, 37].

2.1.2.2 Cellular components

The primary cell types in the aortic valve are: VECs, which form an outer monolayer lining the

surface of the leaflet, and VICs, which are found ubiquitously throughout the leaflet. VECs are

most likely indirectly involved in maintaining valve homeostasis and ECM remodeling by

regulating permeability and adhesiveness to inflammatory cells, and interacting with circulating

cells and local VICs through paracrine signaling [38]. VECs demonstrate phenotypes that are

side-specific [39] and different from vascular endothelial cells [22].

VICs are a heterogeneous population of fibroblasts, with less than 5% as myofibroblasts and

smooth muscle cells [23, 40-42]. Adult human VICs in situ are generally quiescent, expressing

low levels of alpha-smooth muscle actin (α-SMA) and matrix metalloproteinases (MMPs) [22,

23]. The main function of VICs is to maintain normal valve structure and function by remodeling

and repairing the ECM. They are strongly attached to and synthesize ECM, expressing matrix-

degrading enzymes and their inhibitors, which remodel collagen as well as other ECM

components [43].

2.2 Calcific aortic valve disease

Calcific aortic valve disease (CAVD) is the most common valve disease in North America and

Europe [1], comprising almost 15,000 of deaths annually in the United States [2]. CAVD covers

a wide spectrum of pathological changes, including aortic valve sclerosis, where the valve leaflet

thickens and stiffens without functional impairment, and the more advanced form of aortic

stenosis, where functional impairment is present. Currently, CAVD occurs in over 25% of the

population over the age of 65 years and its prevalence is rising [3, 4]. Moreover, it is associated

with many negative clinical outcomes, including a 40% increased risk of myocardial infarction

and a 50% increased risk of cardiovascular death [4]. Progression from sclerosis to stenosis

affects 9% of elderly patients [5] and is associated with an 80% five-year risk of progression to

heart failure, valve replacement, or death [4]. Currently, the only medical intervention is surgical

replacement of the valve. The disadvantages of invasive surgery, durability of biological

replacement valves, and lifelong anti-coagulant therapy with mechanical replacement valves [6]

Page 18: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

8

warrant an improved understanding of CAVD pathogenesis to satisfy the unmet need for

effective medical treatments.

For decades, CAVD was thought to be a disease caused by general “wear and tear” [44]. Now,

disease progression is recognized as an active process, hallmarked by changes in the

organization, composition and mechanical properties of valve ECM [10]. Specifically, lipid

infiltration, alterations in VIC phenotype, inflammation, and calcification occur, and are

mediated by pathways normally involved in valve development and bone and cartilage

metabolism [4, 10-13, 45]. Lesions and calcification predominantly occur in the fibrosa layer of

valve leaflets [10, 11]. This side-specific pathosusceptibility is not fully understood, but may

give insight into the regulatory mechanisms involved in CAVD progression. These changes

result in thickening and stiffening of the valve leaflets, ultimately leading to obstruction of blood

flow and impaired cardiac function. Much of what is known about CAVD stems from studies of

advanced or late-stage valve disease. In contrast, there remains a large gap in our understanding

of early changes. As such, further study of initial pathological mechanisms will vastly improve

our understanding of whole disease progression.

2.2.1 Histopathology of human CAVD

2.2.1.1 Early CAVD lesions

Valvular lesions likely arise due to endothelial dysfunction and/or disruption from high

mechanical forces or low shear stress [39, 46]. Such disturbed flow occurs on the aortic side of

the leaflet [39, 47, 48] where these lesions predominantly form. This side-specific

pathosusceptibility may provide clues to disease etiology. VECs on the fibrosa side of healthy

valves display a side-specific phenotype that is permissive to calcification, expressing genes that

promote or permit skeletal development and vascular calcification [39]. In addition, the aortic-

side endothelium also expresses compensatory protective mechanisms against inflammation and

lesion initiation. Even following a two-week hypercholesterolemic diet, this protective

endothelial phenotype persists on the fibrosa in pigs [49]. Further lending to this notion of side-

specific pathosusceptibility, using fluorescently tagged low density lipoprotein (LDL), it was

shown that the fibrosa side of the valve has a markedly enhanced potential to bind LDL

compared to the ventricularis side in healthy porcine aortic valves [50].

Page 19: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

9

Early human CAVD lesions are characterized by focal subendothelial thickening on the fibrosa

side of the leaflet. The most easily identifiable feature of these lesions is displacement of the

elastic lamina, which often appears fragmented and/or reduplicated [17]. The basement

membrane beneath the endothelium also often appears thin, frayed reduplicated and/or absent

[11]. Forming between the endothelial layer and displaced elastic lamina, early lesions

accumulate protein, lipid, inflammatory cell infiltrate, and extracellular mineralization [11, 17].

While variable levels of collagen and elastin accumulate in early CAVD lesions [11, 17], there is

an overall increase in the three major valve PGs [16]. Localization patterns suggest defined roles

for specific PGs, as biglycan and decorin tend to be present in areas where versican is absent

[16].

Neutral lipid accumulation in early human lesions has been detected by oil red O staining [11,

17, 51]. In contrast to histologically normal regions, lesions and their adjacent fibrosa

accumulate apolipoprotein (Apo) B, Apo(a), and ApoE [17]. Spatial comparisons suggest that

most of the extracellular neutral lipids are related either to plasma-derived or locally produced

apolipoproteins. Since LDL has been observed in early lesions [52], the similar distribution

patterns of ApoB and Apo(a) [17] suggest that both LDL and lipoprotein(a) are deposited. ApoE,

which is largely present in atherosclerotic lesions, is often found in valvular regions with ApoB

and Apo(a), but is also present extracellularly, without ApoB and within macrophages. Since

macrophages increase their secretion of ApoE in response to intracellular cholesterol loading,

much of this ApoE may be produced locally [17].

In early lesions, the presence of inflammatory cells is highly variable with macrophages in only

25-65% of lesions [12, 53] and T-lymphocytes less often [11]. When present, macrophages are

commonly located near the surface of the lesion, while the earliest forms of calcification occur in

a stippled pattern deeper near the base [11, 17]. Importantly, however, early lesions can form in

the absence of macrophages, T-lymphocytes, and calcification [53]. The layered appearance of

lesion features supports the active progression of disease pathogenesis.

2.2.1.2 Advanced CAVD lesions

As CAVD progresses, features observed in early disease, including loss of elastic lamina, protein

accumulation, lipid accumulation, and inflammation, are more pronounced, leading to more

marked thickening of the fibrosa and active tissue calcification.

Page 20: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

10

2.2.1.2.1 Lipid accumulation

Areas of lipid accumulation have also been identified by oil red O staining in late-stage diseased

valves [11, 17, 51], as well as the presence of ApoB, Apo(a), and ApoE [17]. Since ApoB is

present in the absence of Apo(a), the distribution of which is more restricted to the central

regions of the fibrosa, it is likely that both LDL and Lp(a) are deposited.

Oxidized LDL (oxLDL) is also present in stenotic aortic valves, localizing to subendothelial

regions on the fibrosa side with calcium deposition and co-localizing with ApoB and neutral

lipid [51, 54, 55]. OxLDL is both cytotoxic and can stimulate inflammatory activity. Specifically

in the valve, it has been shown to stimulate calcification in valve fibroblasts in vitro [56].

Stenotic valves with high oxLDL scores also have a significantly higher density of leukocytes,

macrophages, and T-lymphocytes compared to valves with lower oxLDL scores [54]. As well, in

diseased valves oxLDL co-localizes with lipoprotein lipase (LPL), which itself localizes in cell

dense areas with abundant macrophages and is associated with valve tissue remodeling.

Sequestration of lipids and oxidative transformation may initiate recruitment of inflammatory

cells, which may further promote the retention of lipids in a cyclic manner [55]. Moreover,

stimulation of aortic VICs with oxLDL has been shown to increase phosphate inorganic

transporter 1 (Pit1) and bone morphogenic protein 2 (BMP2), indicating a potential role for

lipids in osteogenic VIC differentiation and calcification [57].

2.2.1.2.2 Inflammatory processes

In advanced CAVD lesions, an increase in cellularity, including non-foam cell macrophages,

foam cell macrophages, and T-lymphocytes, is present [11, 17, 51, 58-64]. Macrophages are

present in 59-75% [11, 60] and lymphocytes in 75-90% [11, 65] of stenotic valves. Macrophages

and T-lymphocytes are commonly observed near calcium deposits in the subendothelium of the

fibrosa layer [58, 66], co-localizing with areas of neutral and oxidized lipid [51]. Mast cells [62,

67, 68] and B-lymphocytes [67] are also present in stenotic valves. In addition, the presence of a

variety of inflammatory mediators indicates active inflammatory processes. Overall, there is an

increased expression of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNFα)

[51, 63, 64], interleukin-1 [63] and -2 [58], matrix metalloproteinases (MMP-1, -2, -3, -9, -12)

[61, 63, 66, 69, 70] and complement activation [71, 72]. Oxidative stress may also play a role, as

Page 21: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

11

gradients of superoxide and hydrogen peroxide are present with the highest levels occurring in

and near calcified areas [73].

The infiltration of inflammatory cells may create a pro-fibrotic environment involving the renin-

angiotensin system [74]. Increased levels of angiotensin-converting enzyme (ACE) [62, 75],

mast cell cathepsin G and chymase [62], and their enzymatic product, angiotensin II (AngII)

[75], are present. Interestingly, angiotensin II type 1 receptor (AT-1R) is only expressed by valve

fibroblasts in lesions [62, 75], suggesting the reaction of fibroblasts to angiotensin II is blunted

until they express this major pathogenic receptor for AngII [10]. The activation of AT-1R can

increase the production of oxidants and PGs [29, 75].

Endothelial cells also show evidence of inflammatory processes. Adhesion between endothelial

cells and circulating leukocytes is a key initial event in recruiting and transmigrating leukocytes

to sites of inflammation. Those found in valves with CAVD have increased levels of vascular

cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1) [76, 77], and

endothelial selectin (E-selectin) compared to healthy valves [76]. In addition, diseased patients

exhibit elevated levels of E-selectin [76], which is thought to reflect systemic inflammatory

conditions.

2.2.1.2.3 Extracellular matrix remodeling

In advanced disease, the ECM experiences continued remodeling [78]. Similar to early disease

histopathology, distributions of the three major valve PGs, biglycan, decorin and versican, and

the GAG hyaluronan are altered in stenotic valves. In the main report published to date, stenotic

aortic valves removed during valve replacement surgery were immunostained for PGs/GAGs and

analyzed, with focus on expression in and around calcified nodules that were categorized as large

or small, which the authors interpreted as mature or early stage, respectively [14]. Biglycan and

decorin expression within larger calcified nodules was lower than in the regions immediately

surrounding the larger nodules, the regions within and surrounding smaller “pre-nodules”, and

the normal fibrosa, but there were no other significant differences in expression levels of

biglycan or decorin (Fig. 2.2). Versican and hyaluronan expression was lower within larger

nodules than in the immediate surrounding regions and lower within smaller “pre-nodules” than

in the regions surrounding larger nodules, but otherwise not significantly different regionally.

Page 22: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

12

Figure 2.2. Proteoglycan and glycosaminoglycan staining intensities within and surrounding

calcified nodules. p<0.05 compared to Nod Surr. *p<0.05 compared to Fibrosa.

ǂp<0.05 compared to

Prenod. Nod Ctr=innermost 1/3 of the large nodule. Nod Edge=outer 1/3 of the large nodule. Nod

Surr=tissue immediately surrounding the large nodule. Prenod=prenodule. Prenod Surr=tissue

immediately surrounding the prenodule. Error bars = SEM. Adapted with permission from reference [14]

© Elsevier.

Changes in collagen composition and localization also occur, lending to the fibrotic

characteristics observed in advanced CAVD [77]. Dense fibrotic areas are present with

disorganization of collagen fibers in the fibrosa layer. In addition, there is an increase in the

synthesis of type I procollagen, the precursor of the major collagen component of normal valves,

but total collagen is significantly decreased in calcified valve leaflets compared to healthy

control valve leaflets. This suggests that throughout the entire valve there is an overall increased

turnover of type I collagen in stenosis, where degradation exceeds production [79]. Both

collagen II [13, 78] and X [78] are also observed in human adult CAVD, supporting the role of

active osteochondrogenic mechanisms underlying calcification in CAVD. In particular, localized

areas of collagen X expression are observed close to heavily calcified areas of the valve [13].

The disorganization of collagen bundles can also be attributed to the increased presence of

MMPs [69] and their tissue inhibitors (TIMPs) [63, 69]. Fragmented and disorganized elastin

Page 23: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

13

fibers are reported in areas of prominent calcification and initial mineral deposition. This may be

due to increases in elastolytic enzymes, such as MMP-12, which has been detected in its active

form in these areas of ECM alteration, initial mineral deposition, and calcification. Areas of

collagen and elastin fragmentation are both suggested to be potential nidi for calcium deposition

[66].

2.2.1.2.4 Calcification

Ultimately, the valve tissue reaches a stage where it forms calcified nodules, primarily made of

amorphous calcium phosphate [67, 80, 81], and transforms from its natural pliant state to one

that is more rigid and unable to close properly. Calcification co-localizes with areas of lipid

deposition [11, 51] deeper within the lesion [11], and tends to be absent in areas devoid of lipid

[51].

In stenotic valves, calcific nodules are commonly found co-localizing with vasculature [66, 67,

77, 82]. Due to their thinness, diffusion of oxygen from the surfaces of healthy aortic valve

leaflets is sufficient to support the valve’s metabolic needs. When present, which occurs

sparsely, microvasculature is typically found at the base of the valve cusp and within the

ventricularis and spongiosa layers [83, 84]. The presence of neoangiogenesis co-localized with

calcification in diseased states suggests that it may also aid in endochondral ossification [67].

2.2.2 Osteochondrogenic VIC changes

While dystrophic calcification seems to be the major mechanism in heavily calcified valves [85],

heterotopic bone formation also contributes to local valvular calcification [67], indicating that

both passive and active processes are at work. Thirteen percent of calcified aortic valves

demonstrate mature lamellar bone formation with hematopoetic elements and active bone

remodeling, where there is both osteoblastic bone formation and osteoclastic bone resorption

[67]. Earlier stages of osteogenesis are also observed in the form of endochondral bone

formation [66, 67, 86], the process by which long bones are created from the replacement of

bone matrix over a cartilage template. In addition to the presence of bone itself, cartilage is

observed in human stenotic valves independently and with bone [86]. In endochondral

osteogenesis, chondrocytes differentiate from MSCs and as they proliferate, produce collagen

type II, IX and XI, and sulfated GAGs. Further differentiation of the chondrocytes results in

Page 24: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

14

hypertrophic cells that produce collagen type X. The ECM eventually mineralizes, chondrocytes

apoptose, and the calcified cartilage template is infiltrated by capillaries and replaced by bone

[87]. These areas of endochondral ossification in the valve have been described as the

transformation of cartilage into lamellar bone through a zone of provisional calcification [86].

Consistent with this notion of bone formation, several chondrogenic and osteogenic transcription

factors are upregulated in human stenotic valves [88] (Table 2.1). Interestingly, along with their

involvement in cartilage and/or bone formation, many of these transcription factors are also

involved in valve development [13]. Non-calcific, pediatric diseased valves, which may

represent an earlier disease stage, demonstrate an upregulation of chondrogenic pathways with

increases in sex determining region Y box 9 (Sox9), myocyte enhancer factor 2C (Mef2c),

Twist-related protein 1 (Twist1), and muscle segment homeobox 2 (Msx2) [13]. In calcified

adult diseased valves, these chondrogenic transcription factors are also upregulated [13], along

with several other osteogenic transcription factors, including Runt-related transcription factor 2

(Runx2) [13, 80, 89], phosphorylated Smads (p-Smad) 1/5/8 [13], nuclear factor of activated T-

cells cytoplasmic 1 (NFATc1) [89], and osterix (Osx) [89].

Of note to studies of early disease, Sox9-expressing cells were observed in early lesions in a

porcine model of CAVD [19]. Sox9 is important for the differentiation of the chondrocyte

lineage and for the expression of genes characteristic of cartilage, such as collagen 2a1 (Col2a1)

[88, 90, 91]. In teratomas derived from homozygous Sox9 mutant embryonic stem cells, no

cartilage forms although the usual tissue type varieties for teratomas are present, indicating that

the transcription factor plays an essential role in chondrogenesis [92]. Bone morphogenic protein

2 (BMP-2), which is also detected in human stenotic valves [67, 93], increases the expression of

Sox9, as well as its downstream response genes such as Col2a1 [94]. Other mediators of Sox9

upregulation include fibroblast growth factors [95] and hedgehog signaling pathways [94].

Page 25: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

15

Table 2.1. Transcription factors upregulated in CAVD and their roles in chondrogenic and/or

osteogenic processes

Transcription Factor Role in chondrogenesis and/or osteogenesis References

Sox9 Chondrocyte lineage differentiation

Expression of ECM genes characteristic of cartilage

[90], [91]

Mef2c Chondrocyte maturation

Bone formation

[96]

Twist1 Osteoblast differentiation inhibitor and chondrogenesis promoter in

osteoblast progenitors

[97]

Msx2 Proliferation of osteogenic progenitor cells

Bone and cartilage formation

[98], [99]

Runx2 Maturation of osteoblasts

Regulation of osteogenic ECM genes (e.g. collagen X and

osteocalcin)

[100], [101]

NFATc1 Regulation of osteoclast differentiation

Osteoblastic bone formation

[102], [103]

Osterix Osteoblast differentiation

Osteocalcin expression

Bone formation

[104]

In addition, the early CAVD porcine model by Sider et al. positively correlated the expression of

Sox9 with soft PG-rich matrices [18, 19]. In other tissues, expression of Sox9 is stiffness-

dependent and is linked to proteoglycan accumulation. In response to TGF-β1 stimulation,

murine chondrocytes express Sox9 and Col2a1 in a stiffness-sensitive manner [105]. In the

absence of biochemical factors, MSCs cultured on soft substrates (~1 kPa) express higher mRNA

levels of Sox9 and Col2a1 and accumulate PGs in cell aggregates compared to on stiffer

substrates (~15-150 kPa) [106]. As well, the trio of Sox5, Sox6, and Sox9 induces production of

a PG-rich matrix by MSCs [107]. Whether there is a causal relationship between PGs and

osteochondrogenic processes though has yet to be elucidated in valve disease.

2.2.3 Proteoglycans in calcific aortic valve disease

Recently, changes in the abundance and distribution of PG/GAG content in valve leaflets have

garnered interest. In healthy valve tissue, the most common PGs/GAGs are a major component

of the middle spongiosa layer. In advanced CAVD, increases in the PGs biglycan, decorin and

versican, and the non-sulfated GAG hyaluronan are observed in the area immediately

Page 26: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

16

surrounding calcified nodules in the fibrosa layer [14]. Studies of early and advanced CAVD

indicate they may play a crucial role in disease progression.

The formation of PG-rich lesions, similar to those seen in humans [11, 16], are observed in

porcine [18] and mouse (unpublished data) diet-induced models of CAVD. In our swine model

by Sider et al. [18, 19] and in human valves with early disease [11, 17], these PG lesions are

observed before the appearance of myofibroblasts, macrophages, dendritic cells, and significant

lipid accumulation. I corroborated the absence of myofibroblasts by both immunoperoxidase and

immunofluorescence methods in the swine model (Appendix A.1). This suggests that PG lesion

formation is an initial step in CAVD pathogenesis, occurring before VIC activation,

inflammation and lipid retention. Further, early PG-rich lesions tend to be softer compared to

normal fibrosa, which may be more permissive to chondrogenesis. As discussed above,

expression of chondrogenic markers appears to be stiffness-dependent [105, 106]. Putative

chondrogenesis is supported by the increased presence of Sox9-positive cells observed in early

porcine CAVD lesions [18].

2.2.3.1 Implications of the role of proteoglycans from atherosclerosis

CAVD shares several risk factors with atherosclerosis, including hypertension,

hypercholesterolemia, smoking, male gender, diabetes, chronic renal disease and older age [3,

11]. Consequently, it is believed CAVD may have similar pathobiological processes as

atherosclerosis. In early lesions, characteristics shared by both diseases include displaced elastic

lamina, lipid infiltration and involvement of inflammatory cells [11]. Furthermore, the initiating

factor for progression to CAVD and atherosclerosis seems to involve endothelial injury at sites

of low shear and high tensile stress.

Although the role of PGs/GAGs in CAVD has not been extensively studied, it is suspected they

may aid in the initiation of disease by retaining lipids and binding macrophages, as they do in

atherosclerosis [15, 108]. Although recognized by the Council of American Heart Association as

normal intima, diffuse intimal thickening (DIT), which consists of PGs, elastin and smooth

muscle cells (SMCs), is thought by some to be a precursor of atherogenesis, as it consistently

presents in atherosclerosis-prone arteries and not in atherosclerosis-resistant arteries [108, 109].

The predominant PGs in these atherosclerosis-prone arteries are biglycan and versican, while

Page 27: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

17

atherosclerosis-resistant arteries are thin and enriched in decorin [110, 111]. Decorin follows a

similar distribution pattern to biglycan in DIT, but has far fewer positively stained areas [109].

DIT occurs before lipoprotein deposition [15] with only a small number of macrophages present

and with no evidence of neovascularization [108]. The response-to-retention hypothesis suggests

that a predisposing factor, such as mechanical stress or cytokines, stimulates the local synthesis

of PGs that have a high binding affinity for lipoproteins [108]. Once these atherogenic ApoB-

containing lipoproteins enter the arterial intima, it is thought that they are retained by PGs [112].

The resulting lipoprotein-PG complexes are more susceptible to modifications, such as oxidation

and aggregation, which lead to uptake by macrophages to form foam cells. As well, the resulting

oxidized lipids may promote further production of PGs that have a high affinity to lipoproteins

[15].

In atherosclerosis, PGs bind lipoproteins through ionic interaction via their negatively charged

GAG chains, which can be mediated by accessory molecules such as lipoprotein lipase (LPL)

[15, 113-115]. The lipid-binding capacity of PGs, which relates to GAG chain length and

sulfation, contributes to the retention of lipoproteins in the intima [116-119]. The most common

PG in the vascular ECM is versican, followed by biglycan and decorin. In vitro studies have

shown that of the three major PGs, versican has the greatest potential to bind lipoproteins

because of its high number of LDL binding sites [111, 113]. In contrast, in vivo studies show that

biglycan and decorin most commonly co-localize with LDL in early atherosclerotic lesions [15,

112]. Overexpression of human biglycan by rat SMCs results in production of an ECM with

greater high-affinity lipoprotein binding [120]. Furthermore, pre-lesion biglycan was localized in

a similar distribution to lipids in the early phase of atherosclerotic lesions [15], suggesting that

biglycan may play an important role in the very initial stages of lipid deposition. Decorin may

play a role in linking lipoproteins to collagen in atherosclerosis, as it has been shown to link LDL

with collagen type I in vitro and co-localize with collagen and ApoB in atherosclerotic lesions in

vivo [108]. A transgenic mouse model of human ApoB100, which expressed PG-binding

defective LDL, receptor-binding defective LDL, or wild-type LDL further validated the

importance of PG binding to LDL in the initial stages of atherosclerosis [121]. Mice fed an

atherogenic diet were less susceptible to atherosclerotic lesion formation if they had PG-binding

defective LDL instead of wild-type LDL. Moreover, mice with different LDLs demonstrated no

Page 28: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

18

difference in susceptibility to oxidation and macrophage uptake. Therefore, reduced

atherogenesis was likely due to reduced PG-binding ability.

Despite certain similarities between atherosclerosis and CAVD, implications drawn from

atherosclerosis research must be taken with a grain of salt. Less than 40% of patients with

CAVD have clinically significant coronary atherosclerosis [122]. In addition, although statins are

widely used to effectively treat atherosclerosis, randomized controlled studies have yet to show

their effectiveness in the treatment of valve disease [7-9]. Evidently, some distinct processes are

involved in CAVD. In contrast to atherosclerosis, there is no SMC involvement in CAVD [11].

As well, since lipoproteins are observed in the adjacent fibrosa, it is evident that in CAVD, early

lesions are not confined to the area bound by the elastic lamina, as they are in vascular disease.

Although some insight into CAVD progression may arise from studies of atherosclerosis,

differences between the diseases warrant further individual study of CAVD progression.

2.2.3.2 Localization and function of specific PGs/GAGs in CAVD

PGs/GAGs exhibit spatial heterogeneity in both early and advanced CAVD lesions. In early

lesions, versican tends to be absent in areas with biglycan and decorin [16]. Surrounding and

within calcified nodules in the fibrosa layer of stenotic valves, spatially heterogeneous changes

in the major valve PGs, as well as hyaluronan, are observed with dependence on nodule size [14]

(Figure 2.2). In stenotic leaflets, PGs and hyaluronan were found to have the greatest expression

in areas directly surrounding calcified nodules. Within smaller calcified nodules, termed

“prenodules” and interpreted to be less advanced, and surrounding regions, decorin and biglycan

are significantly more abundant compared to within and surrounding larger calcified nodules.

Biglycan and decorin may accumulate in early nodule formation, but as the nodule becomes

more mineralized, be more involved in remodeling the tissue surrounding nodules. Within

prenodules, versican and hyaluronan are negatively correlated with biglycan and decorin,

suggesting they may be less involved in early nodule formation. Their presence surrounding

nodules and prenodules suggests they may be more involved in remodeling areas surrounding

mineralized tissue.

Insight into the localization of specific PGs/GAGs involved in early CAVD will aid the

understanding of the pathobiological changes that result in calcification and stenosis.

Proteoglycan function and categorization is largely determined by the composition of its GAG

Page 29: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

19

chains, which can differ in sulfation pattern, disaccharide composition, and chain length [29].

Decorin and biglycan are both small leucine-rich PGs that are composed of one and two GAG

chains, respectively, from chondroitin sulfate and/or dermatan sulfate. Versican is a large

chondroitin sulfate PG, which occurs in four isoforms: V0, V1, V2, and V3. Hyaluronan is a

non-sulfated GAG and instead, interactions occur largely through receptors such as CD44,

receptor for hyaluronan-mediated motility (RHAMM or CD168), and hyaluronan receptor for

endocytosis (HARE).

Proteoglycans potentially play a direct and indirect role in lipid retention during the early stages

of CAVD. Glycosaminoglycan chains contain negatively charged sulfate groups and carboxylic

groups, which allow PGs to interact with positively charged lysine and arginine residues, such as

those on apolipoproteins. Multiple LDL particles can bind to a single GAG chain [123, 124]. Of

the major valve PGs, decorin has one GAG chain, biglycan has two GAG chains, and versican

isoforms range up to 23 GAG chains [125]. In valve lesions though, apolipoproteins have been

observed to co-localize with biglycan and decorin [16, 17]. Using LDL affinity columns, it was

shown that decorin and biglycan are major mediators of lipid retention in porcine aortic valves

[50]. Structural properties of GAG chains that may influence apolipoprotein binding include

GAG chain length and sulfation pattern. In vitro studies demonstrate that PG binding affinity for

LDL is augmented with increasing GAG chain length [126]. Interestingly, GAG chains bound to

PG core proteins show higher affinity binding to LDL compared to free chains due to

thermodynamic considerations of molecular rigidity [117]. Several factors have been shown to

cause elongation of chondroitin sulfate chains, including TGFβ and oxLDL [127]. Subtle

changes in sulfation are also thought to be able to alter the ionic interactions of GAGs with

apolipoproteins. For example, in comparison to 4-sulfated GAGs, 6-sulfated GAGs are more

sterically accessible to the binding sites on LDL [29]. These PG-LDL interactions are largely

electrostatic in nature, as denaturing agents, SDS, and urea resulted in little, if any, eluent

following salt elution. Further, proteoglycans, specifically decorin, can act as bridging molecules

to mediate LDL-collagen interactions [50]. In addition to direct ionic interactions with lipids,

proteoglycan GAG chains are able to bind lipoproteins via bridging molecules, including LPL

[126] and ApoE [128].

Lipid retention then allows modifications of lipoproteins by enzymes, such as, hepatic lipase

(LIPC), phospholipid transfer protein (PLTP) and LPL [112], which can result in further lipid

Page 30: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

20

retention and inflammation. In valve disease, biglycan induces increased expression of

phospholipid transfer protein (PLTP) by VICs via Toll-like receptor 2 (TLR2) [129] and decorin

has been shown to co-localize with LPL [55]. In these studies, biglycan and decorin were also

found to co-localize with oxLDL, which is associated with inflammation [54] and has been

implicated in osteogenic VIC differentiation [57]. Interestingly, biglycan has been shown to

contribute to the pro-osteogenic effect of oxLDL on human aortic VICs [130]. Stimulation of

VICs with oxLDL increases expression of biglycan, which in its soluble form can induce the

expression of BMP2 and ALP via TLR2.

The small leucine-rich PGs biglycan and decorin are also known to mediate collagen

fibrillogenesis [14, 131] and sequester TGF-β [14, 34]. The latter is particularly significant, as

TGF-β1 has been shown to induce stiffness-dependent VIC differentiation to chondrogenic or

myofibroblastic phenotypes [37, 132, 133] and stimulate expression of MMPs [134], which

mediate further ECM remodeling in disease. In addition, TGF-β1 has been shown to induce PG

core protein synthesis and GAG chain elongation in porcine VICs. Interestingly, PGs synthesized

in response to TGF-β1 demonstrate enhanced LDL binding [126].

Studies involving hyaluronan thus far demonstrate both potential protective and pathogenic roles

in the aortic valve. In stenotic human aortic valves, hyaluronan abundance is inversely related to

the magnitude of observed fibrosa layer calcification [135]. As well, VICs treated with

hyaluronan have suppressed calcified nodule formation, indicating it may have a protective role

[21]. This is consistent with other cell types, where hyaluronan attenuates the cellular response to

TGF-β1 [136]. In atherosclerosis though, hyaluronan has the ability to retain lipids [137] and is

involved in the accumulation and activation of inflammatory cells [138, 139], indicating it may

also play a role in lipid retention and chronic inflammation during CAVD. The presence of

hyaluronidase-1 and hyaluronan synthases co-localized with differentiation markers of brown fat

cells and chondrogenesis within and surrounding calcified nodules suggests that turnover of this

GAG has a role in disease progression [140].

The role of PGs in mineralization processes is not clear, but their GAG components, particularly

chondroitin-4-sulfates, have the capacity to bind calcium and interact with hydroxyapatite [141].

While the direct pathobiological roles of PGs/GAG in valve calcification have not yet been fully

explored, it is suspected that they may aid in lipid retention, which may initiate a cascade of

Page 31: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

21

inflammation, osteogenesis and/or apoptosis, leading to calcification. Overall, an improved

understanding of the changes that occur in the ECM, particularly specific PGs/GAGs, as well as

the effect these changes in lesion microenvironment have on VICs, is required to define their

contribution to early CAVD progression.

2.2.4 Porcine models of calcific aortic valve disease

Due to the availability of human stenotic valves upon valve replacement, late-stage CAVD is

well-characterized. Knowledge of the initiating events involved in CAVD has been limited

though by the difficulty of retrieving suitable samples that represent the desired disease stage and

that are controlled for by confounding factors in human autopsy or transplant patients.

Consequently, the use of animal models is necessary to satisfy the unmet need to uncover the

pathobiological processes involved in CAVD.

The animals most commonly used in the study of CAVD are mouse, rabbit, and swine [142].

Swine are thought to be excellent models for the study of atherosclerosis [143, 144] and recently,

of CAVD [142, 145] because of their many similarities to humans. Unlike mice, swine have tri-

layered aortic valve leaflets, which is an important feature considering the pathosuceptibility of

the fibrosa side to forming CAVD lesions. Swine also spontaneously develop atherosclerotic,

human-like lesions, a process that is accelerated by high-fat/high-cholesterol diets [146]. In

contrast, wild-type mice fed standard diets do not exhibit spontaneous calcification [145] and

usually require diet and/or genetic predisposition to induce advanced disease [147-150]. Rabbits

also do not develop spontaneous atherosclerotic lesions and have a significantly different lipid

metabolism compared to humans [143]. As a result, they usually require very high cholesterol

levels to induce advanced disease [151-153]. When fed an atherogenic diet, swine exhibit similar

lipid profiles [154] and lipoprotein metabolism [146, 155] as humans. Furthermore, swine and

human genomes are comparable in size and homologous in sequence and chromosomal structure,

making porcine models useful for genomic studies [145]. Swine have not been extensively used

in CAVD research though, mainly due to their large size, which creates limitations because of

cost and maintenance issues.

Studies of CAVD in swine show that they develop human-like lesions when fed a

hypercholesterolemic diet [18, 39, 49]. Distinct areas of subendothelial lipid accumulation and

early calcific nodules are present on the fibrosa side at two weeks and moreso, at six months [39,

Page 32: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

22

49]. At both time points, no frank inflammation is present [49]. This diet also induces a side-

specific protective phenotype in fibrosa side VECs that is anti-calcific, anti-apoptotic, and anti-

inflammatory.

2.2.4.1 A porcine model of early calcific aortic valve disease

Recently, a porcine model was developed by Sider et al. to gain further insights into the early

ECM changes that occur with CAVD [18, 19]. This porcine model successfully mimics many

characteristics of early human CAVD, which are enhanced by hypercholesterolemia. Moreover,

the diets achieved cholesterol levels for both control and experimental groups that are

comparable to normal humans and those with familial hypercholesterolemia, respectively.

The fibrosa side of these valve leaflets forms human-like early CAVD lesions, the most

advanced of which develop after being fed HF/HC diet for 2 or 5 months. Lesions are composed

primarily of PGs with varying amounts of collagen and elastin, which are laid down between the

endothelial cell layer and the displaced, fragmented and/or reduplicated elastic lamina on the

fibrosa side of the valve. Often, these changes in the elastic lamina are the first visible sign that

there is disruption of the normal valve microstructure. These lesions occur in the absence of

myofibroblasts, osteoblasts, macrophages, and dendritic cells, indicating that they, indeed,

represent an early stage of valve disease.

Lesions from pigs fed the HF/HC diet also have a greater presence of ApoB, Sox9-positive cells,

and Msx2-positive cells compared with pigs fed normal chow, suggesting a role in lipid retention

and putative osteochondrogenesis. Although it has been proposed that lipid retention is an initial

step of valve disease, ApoB was present in only 28% of all lesions, suggesting that it is preceded

by PG deposition. According to layer-specific stiffness analysis using micropipette aspiration,

these early lesions also have a tendency to be softer than normal fibrosa. In addition, soft onlays

contained more PG and less fibrillar collagen compared to normal fibrosa. It has been suggested

that this soft, PG-rich microenvironment is permissive to increased Sox9 expression by resident

VICs, but this causal relationship has yet to be elucidated.

An important limitation of this model is that no calcification was observed up to the time points

studied. Although markers associated with early osteogenic differentiation were present, this

does not definitively support initiation of mineralization. Still, diabetic swine have been shown

Page 33: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

23

to develop early limited valvular calcification over similar time periods [39, 49] and other swine

on an atherogenic diet formed atherosclerotic lesions with calcification [146], indicating the

potential for cardiovascular calcification in swine. Nevertheless, the similarities with early

human disease suggest that this porcine model allows for critical insights into the initial stages of

lesion formation and sclerosis.

Page 34: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

24

Chapter 3

3 Hypotheses and Objectives

3.1 Hypotheses

There are temporal changes in the amount of specific proteoglycans and glycosaminoglycans

(PGs/GAGs) in early PG-rich lesions as calcific aortic valve disease (CAVD) progresses. In

particular, it is hypothesized that levels of biglycan, decorin, and versican increase and

hyaluronan decrease with increasing time and administration of a high-fat/high-cholesterol diet.

Further, it is hypothesized that PGs/GAG found in more advanced lesions associate with lipid

retention and Sox9-expressing cells. With these changes in the lesion microenvironment, there

are phenotypic differences between valve interstitial cells (VICs) in lesions versus the normal

fibrosa.

3.2 Objectives

Using a porcine model of early CAVD:

(1) To quantify the amount of specific PGs/GAG in early CAVD lesions using

immunohistochemistry;

(2) To define correlations between PG/GAG content in early CAVD lesions with lipid

retention and Sox9-expressing cells using immunohistochemistry; and

(3) To characterize the phenotypic differences between VICs in the lesion and healthy

fibrosa using microarrays.

Page 35: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

25

Chapter 4

4 Proteoglycan and glycosaminoglycan content in lesions of early CAVD

4.1 Introduction

Once thought to be a disease of passive degeneration [44], calcific aortic valve disease (CAVD)

is currently recognized as an active process in which cellular and extracellular matrix (ECM)

changes result in thickening and stiffening of the valve leaflets, which ultimately lead to

obstruction of blood flow and impaired cardiac function [10]. Due to the availability of suitable

tissue samples from aortic valve replacement, advanced disease has been extensively studied and

is characterized by ECM disorganization, fibrosis, and calcification [10, 67]. Still, CAVD

persists as a prevalent disease with poor clinical consequences and no effective medical therapy

[3, 4]. Consequently, studies of early disease processes are essential to better understand disease

progression and to find novel targets for CAVD treatment.

Aortic valve leaflets demonstrate a side-specific pathosusceptibility, preferentially forming

CAVD lesions on the fibrosa side of the valve. Early lesions are characterized by areas of focal

subendothelial thickening with displacement of the elastic lamina, which often appears

fragmented and/or reduplicated, and a thin, frayed, reduplicated, or absent basement membrane

[11, 17]. This subendothelial thickening often accumulates lipid, inflammatory cell infiltrate,

extracellular mineralization, and protein [11, 17]. Lipoproteins are commonly found within these

lesions [11, 17] and often co-localize with PGs [16, 52]. The presence of macrophages in these

early lesions tends to vary drastically from none to substantial [11, 53], but when present, occur

near the surface of the lesion or in a stippled pattern near the base of the earliest calcified lesion

[11, 17]. This layered appearance further supports the notion of the active progression of CAVD

[11].

Early studies of CAVD have been hampered by the lack of suitable human tissue samples and

well-characterized animal models. Recently though, studies of early CAVD in swine showed that

they develop human-like lesions when fed an atherogenic diet [18, 39, 49]. Pigs are considered to

be excellent models for studies of atherosclerosis [143, 144], and recently, of CAVD [142, 145].

Page 36: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

26

Compared to humans, they have (1) similar lipid profiles [154] and metabolism [146, 155]; (2)

comparable genome size and homology [145]; and (3) develop atherosclerotic-like lesions with

age, which is accelerated with HF/HC diet [146].

The porcine model developed by Sider et al. [18, 19] is able to successfully mimic several

characteristics of early CAVD lesions seen in humans [11, 17]. These lesions form before the

appearance of myofibroblasts, significant lipid accumulation, significant inflammatory cell

infiltrate, and calcification, indicating that they represent a very early disease stage. Interestingly,

while collagen and elastin content varies, PGs appear as the primary component of accumulation

within these lesions. Proteoglycans are typically found in the spongiosa layer of healthy aortic

valves, but accumulations have been observed in both early sclerotic [16] and advanced stenotic

valves [14]. From this porcine model of early CAVD, it is suggested that PG accumulation

precedes lipoprotein retention, since the formation of PG-rich lesions often occurs without any

detectable lipoprotein deposition. Nevertheless, ApoB deposition and Sox9 and Msx2 expression

scores are greater in PG-rich lesion than non-lesion areas, suggesting a role for PGs in lipid

retention and putative osteochondrogenesis.

In advanced CAVD, increases in the PGs biglycan, decorin, and versican, and the non-sulfated

GAG hyaluronan have been observed in the area immediately surrounding calcified nodules in

the fibrosa layer [14]. This chapter seeks to elucidate the localization patterns of these specific

PGs/GAGs in the aforementioned early porcine model of CAVD.

4.2 Materials and Methods

4.2.1 Porcine model of early calcific aortic valve disease

The porcine model developed by Sider et al. [18, 19] was used (Appendix A.2). Male Yorkshire

barrows were fed either a normal or high-fat/high-cholesterol (HF/HC) diet for 2- or 5-months

with 6 swine per group. The control grower corn/soybean diet consisted of 15% protein, 75%

carbohydrate, and 10% fat (kcal%) at 3.51 kcal/g. The experimental grower diet consisted of

15% protein, 53% carbohydrate, and 32% fat at 4.02 kcal/g, due to the addition of 12% lard and

1.5% cholesterol. Two-month control and two-month experimental pigs (start weight: 59-65 kg)

were fed the control diet at 3.51 kcal/g and experimental diet at 4.02 kcal/g, respectively, for 65

days. Five-month control pigs (start weight: 14-16 kg) were initially fed a standard

Page 37: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

27

corn/soybean-based starter III diet, due to their young age, containing 19% protein, 71%

carbohydrate, and 10% fat at 3.51 kcal/g until ~40 kg followed with the control grower

corn/soybean diet up until 155 days. Five-month experimental pigs (start weight: 14-16 kg) were

fed the starter III diet supplemented with an additional 12% lard and 1.5% cholesterol,

containing 19% protein, 49% carbohydrate, and 32% fat at 4.01 kcal/g until ~40 kg followed

with the experimental grower diet up until 155 days.

The experimental diet was fed at 87% of the mass of the control diet to provide isocaloric intake.

Feed was adjusted weekly by weight to achieve a growth rate of ~0.75 kg/day (actual average:

0.88 kg/day) for the 2 month pigs and ~0.70 kg/day (actual average: 0.62 kg/day) for the 5 month

pigs. Swine, weighing 106-126 kg, were sacrificed by electric shock and bleed out, according to

standard abattoir practices.

The protocol was approved by the University of Guelph and University of Toronto Animal Care

Committees. All animals were housed individually and treated in accordance with the

recommendations of the Guide for the Care of Laboratory Animals published by the United

States National Institute of Health [156].

4.2.2 Porcine leaflet handling

In total, 24 right coronary leaflets (6 per group) were harvested within 2 hours of sacrifice and

frozen in 10% dimethyl sulfoxide (DMSO) in RPMI medium in a 1°C freezing container and

stored at -180°C. After 6-7 months, leaflets were then thawed for 5 min in a 37°C bath and

DMSO was removed with three 10 min PBS with Ca+2

and Mg+2

doublings on ice. Leaflets were

then fixed in 10% neutral buffered formalin for 48 hours at room temperature and stored in 70%

ethanol at 4°C for 1-2 months. Due to technical difficulties, one 2-month control leaflet was not

processed. Right coronary leaflets were segmented along the circumferential midline, embedded

in paraffin, and cut into 5 µm sections according to routine procedure (Appendix B.1).

4.2.3 Histological and immunohistochemical staining

Radial centre sections of the valve leaflet were examined for PG/GAG content, as they provide a

good representation of whole valve leaflet composition. Sections were stained by Movat’s

pentachrome (Electron Microscopy Sciences, Hatfield, PA, USA) to identify collagen (yellow),

PGs (blue), cytoplasm, muscle (red), elastin, and cell nuclei (black) components (Appendix B.2).

Page 38: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

28

This staining also allowed for visualization of the three valve leaflet layers. Elastin staining by

resorcin-fuchsin (Electron Microscopy Sciences) was also used to highlight reduplication and/or

displacement of the elastic lamina.

Staining for biglycan, decorin, versican, and hyaluronan was performed on serial centre sections

to correlate spatial expression of specific PGs/GAG. In addition ApoB and Sox9 staining was

completed to correlate lipid deposition and an early chondrogenic marker with PG/GAG

expression. Biglycan (anti-biglycan; goat polyclonal, 20 µg/mL, sc-27936, Santa Cruz

Biotechnology Inc., Santa Cruz, CA, USA), decorin (anti-decorin; rabbit polyclonal, 10 µg/mL,

sc-22753, Santa Cruz Biotechnology Inc.), versican (anti-versican; rabbit polyclonal, 1.3 µg/mL,

16770002, Novus Biologicals, Littleton, CO, USA), ApoB (anti-ApoB; sheep polyclonal, 3.6

µg/mL, AHP214, AbD Serotec, Raleigh, NC, USA) and Sox9 (anti-Sox9; rabbit polyclonal, 3

µg/mL, ab26414, Abcam, Cambridge, MA, USA) were detected immunohistochemically, while

hyaluronan was detected using biotinylated hyaluronan-binding protein (bovine nasal cartilage,

1.25 µg/mL, 385911, Calbiochem, Gibbstown, NJ, USA) (Appendix B.3).

Briefly, IHC began by melting the wax on the sections at 60°C for 30 min. This was followed by

deparaffinization in xylene and rehydration in graded ethanol baths. Antigen retrieval for Sox9

staining involved incubation in Tris-EDTA for 30 min in a 98°C water bath, followed by cooling

in room temperature water for 20 min. All other IHC staining protocols utilized antigen retrieval

using 1 µg/µl Trypsin-CaCl2 (Sigma-Aldrich Canada Ltd., Oakville, ON, Canada) for 30 min in

a 37°C water bath. Endogenous peroxidases were then blocked with 3% H2O2/methanol for 10

min. Non-specific staining was blocked using the appropriate 1% serum buffer for 45 min prior

to an hour-long incubation with primary antibody or binding protein. Samples, with the

exception of those for hyaluronan staining, were then incubated with biotin-labeled secondary

(anti-goat, anti-sheep, or anti-rabbit; Vector Laboratories, Burlington, ON, Canada) in 2%

specific serum (rabbit, horse, or goat) for 30 min. All samples were incubated with avidin-biotin-

peroxidase conjugate (Vectastain Elite ABC Kit, Vector Laboratories) for 30 min.

Positive staining was visualized following a 5 min incubation in Vector NovaRED (Vector

Laboratories), followed by Vector Hematoxylin QS (Vector Laboratories) counterstaining.

PBS/Tween was used to wash between steps. For all staining protocols except hyaluronan,

negative controls involved no primary and IgG controls (Santa Cruz Biotechnology Inc. or R&D

Page 39: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

29

Systems, Minneapolis, MN, USA). For hyaluronan staining, negative controls involved no

primary controls and sections pre-treated with 110.25 units/mL hyaluronidase (Streptomyces

hyalurolyticus, H1136, Sigma-Aldrich Canada Ltd.) and stored in PBS at 4°C until use.

4.2.4 Data retrieval and statistical analyses

PG-rich lesions were identified using Movat’s pentachrome. For each lesion, images of the serial

sections stained with Movat’s pentachrome, and for the PGs/GAG of interest, ApoB, Sox9, and

elastin were layered in Adobe Photoshop so that lesion areas overlapped. Displaced elastic

lamina, as identified by elastin staining, was used to trace out and isolate only the lesion area in

each of the layered images.

Quantitative data were retrieved from these lesion images using algorithms in ImageJ, which

calculated total stained area based on hue, saturation, and brightness thresholds (Appendix B.4).

The total area of each lesion was calculated by subtracting white space area from the total image

area. These values were used to calculate percent PG/GAG staining of total lesion area. For the

Movat’s pentachrome images, a similar algorithm was used to separate and quantify: (1)

proteoglycans, (2) collagen, and (3) cytoplasm, elastin, and nuclei. For each lesion, cell density

was quantified. Furthermore, each lesion was qualitatively described as morphologically dense

or diffuse (Figure 4.1). These criteria allowed for characterization of whole lesion areas.

Figure 4.1. Qualitative classification of lesion morphology. (A) Dense and (B) diffuse proteoglycan-

rich lesions stained with Movat’s pentachrome staining (blue = proteoglycans, yellow = collagen, black =

nuclei/elastin, red = cytoplasm). Scale bar = 80 µm.

Localized analysis of lesion areas was performed by dividing each lesion into grids with 100 µm

x 100 µm grid cells. Each grid cell was scored on a semi-quantitative scale based on staining

A B

Page 40: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

30

intensity and area for ApoB and PG/GAG (Figure 4.2). For grid cells with ApoB positive areas,

PG/GAG score was assessed only within the lesion area with positive ApoB staining (i.e., in co-

localized ApoB and PG/GAG areas within the lesion in a grid cell). For grid cells without ApoB

positive areas, PG/GAG score was designated based on the entire lesion area present in the grid

cell. The proportion of Sox9-positive cells was also quantified in each grid cell to elucidate any

relationship between PG/GAG type and putative chondrogenesis. For Sox9 analyses, PG/GAG

scores were assigned based on staining observed in the entire grid cell. Using a grading system

based partially on staining intensity has limitations. For example, technical issues such as section

thickness and staining performed on different days could alter levels of staining intensity. To

validate this PG/GAG scoring system though, scores were normalized and combined for each

lesion and compared to the data for percent PG/GAG staining of total lesion area. These results

strongly correlated for each PG/GAG, suggesting that technical variations in staining intensity

did not have a major influence on scoring (Appendix A.3). Cell density was also measured by

counting all cells in the images stained with Resorcin Fuchsin and dividing the cell count by the

total lesion area.

Due to the small sample size (n=50), the Shapiro-Wilk Test was used to assess the normality of

lesion data sets to determine whether parametric tests or non-parametric equivalents would be

used for analyses. Firstly, percent staining of total lesion area was compared for PG/GAG and

the Movat’s pentachrome separation variables between swine from the different time points and

diet groups using one-way ANOVA with Tukey’s post-hoc test or Kruskal-Wallis Test with

Mann-Whitney post-hoc tests. Bonferroni corrections (significance: p<0.0083) were applied to

these Mann-Whitney post-hoc tests to account for multiple comparisons. Changes in cell density

were also compared between groups using one-way ANOVA followed by Tukey’s post-hoc

tests. Secondly, localized areas from all lesions were characterized using Kruskal-Wallis tests

and Mann-Whitney post-hoc tests to compare PG/GAG score with ApoB score and Sox9

fraction. To account for multiple comparisons, Bonferroni corrections (significance: p<0.005)

were applied to post-hoc tests. Lastly, morphologically dense and diffuse lesions were compared

for PG/GAG composition, ApoB score and Sox9 fraction using Mann-Whitney tests. In addition,

the frequency of morphologically dense and diffuse lesions was compared between different time

points and diet groups using Fisher’s exact test. All statistical analyses were performed on IBM

SPSS Statistics (version 20.0.0) with significance level p<0.05, unless otherwise stated.

Page 41: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

31

0 1 2 3 4

Ap

oB

Big

lyca

n

De

corin

Ve

rsic

an

Hya

luro

na

n

Figure 4.2. Semi-quantative scoring system for ApoB and PG/GAG content. Sample images for each

scoring category (0-4).

Page 42: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

32

4.3 Results

4.3.1 HF/HC diet alters lesion ECM composition with temporal differences

In total, 50 PG-rich lesions were identified with Movat’s pentachrome staining and ranged from

36 to 376 µm in thickness. Analysis of lesion PG/GAG content demonstrated significant

differences in PG composition between the time point and diet groups for each of the PGs/GAG

(p<0.05) according to one-way ANOVA and Kruskal-Wallis (Figure 4.3). Differences, in

particular, were observed with the HF/HC diet compared to normal chow at two months.

Specifically, increases in biglycan (p<0.001) and decreases in hyaluronan (p=0.009) with the

HF/HC diet were noted at this time point. After five months on the HF/HC diet, lesions

expressed less biglycan (p<0.001), decorin (p=0.003) and versican (p<0.001) than at two months.

There were no significant differences based on the Bonferroni-corrected p-value of 0.0083

between the control groups at two months and five months for any of the PGs/GAG (biglycan:

p=0.101, decorin: p=0.010, versican: p=0.017, hyaluronan: p=0.999). These trends involving

biglycan and decorin were recapitulated in analyses of PG/GAG lesion scores (Appendix

A.3).Additionally, while there was apparent variability in the presence of each of the PGs, some

moderate level of hyaluronan remained largely consistent within most lesion areas.

Between the time point and diet groups, for swine fed the HF/HC diet, lesions from 5-month pigs

had increased proportions of collagen (p<0.001) and decreased proportions of nuclei, cytoplasm,

and elastin (p<0.001) compared to lesions from 2-month pigs (Figure 4.4). Overall, cell density

remained consistent between the time point and diet groups, but was less in the 5-month control

versus 5-month experimental lesions (p=0.036).

Page 43: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

33

MP Biglycan Decorin Versican Hyaluronan

s2

Mo

nth

Co

ntr

ol

HF

/HC

5 M

onth

Co

ntr

ol

HF

/HC

Figure 4.3. HF/HC diet altered lesion PG composition with temporal differences. HF/HC = high-fat/high-

cholesterol diet. MP = Movat’s pentachrome (blue = proteoglycan, yellow = collagen, black = nuclei/elastin, red =

cytoplasm/muscle). Error bars = SEM. *p<0.0083

* * *

*

*

Page 44: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

34

Figure 4.4. Temporal changes in lesion ECM composition. Movat’s pentachrome staining revealed

increases in collagen and decreases in “other” (nuclei, cytoplasm, and elastin) between the 2-month and

5-month HF/HC diet lesions. HF/HC = high-fat, high-cholesterol. Error bars = SEM.

4.3.2 Lesion areas that contain ApoB and Sox9-expressing cells display unique PG/GAG composition

ApoB staining was typically subendothelial, but there were a few cases of staining deeper within

the lesion. Lesion areas were semi-quantitatively scored for ApoB and spatially correlated to

PG/GAG content to characterize the relationship between specific PG/GAG presence and lipid

retention in early valve disease. ApoB scores were compared between scores for each PG/GAG

using Kruskal Wallis tests, followed by Mann-Whitney post-hoc tests. For biglycan and decorin,

there was no difference in ApoB score between areas with low scores (0 to 2) of these PGs.

ApoB scores also did not differ between the higher scoring (3 and 4) areas for biglycan

(p=0.378) and decorin (p=0.012). Overall, increases in biglycan and decorin score were

associated with increased ApoB score (p<0.004) (Figure 4.5A,C). Areas with no versican had

higher ApoB scores than areas with versican (p<0.002), but in general, varying versican scores

did not differ in ApoB score (Figure 4.5E). Between all hylauronan scores, there was no

difference in ApoB score (Figure 4.5G).

Page 45: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

35

(A)

(B)

(C)

(D)

(E)

(F)

(G)

(H)

Figure 4.5. Relationship between ApoB and PG/GAG score in lesion areas. Increased apolipoprotein B

(ApoB) severity was associated with higher scores of biglycan and decorin. Error bars = SEM. *p<0.005

Overall, average ApoB scores for all levels of each PG/GAG were low (> 1.7). When the

frequencies of PG/GAG scores were sorted by ApoB score, it was evident that there was

immense variability in PG/GAG scores when there was no ApoB present (Figure 4.5). Still, the

*

*

*

Page 46: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

36

trends observed from Kruskal Wallis tests were visible with the skew in frequencies of PG/GAG

scores with changing ApoB scores.

Sox9-positive cells were often observed near the base of early CAVD lesions (Figure 4.6).

Within lesion areas, the fraction of Sox9-expressing cells and area PG/GAG score were used to

identify the relationship between PG/GAG expression and indications of early chondrogenesis

based on previous findings by Sider et al [19] (Figure 4.7). Overall, there was no difference in

Sox9 fraction between lesion areas with low biglycan scores (0 and 1). Sox9 fraction also did not

differ between the higher scoring (3 and 4) areas for biglycan (p=0.822). Compared to low

biglycan scoring areas, there was an increase in fraction of Sox9-positive cells within high

biglycan scoring areas (p<0.002). For decorin and versican, there were largely no differences in

proportion of Sox9-positive cells between PG scores. With higher hyaluronan score, there was a

decrease in Sox9-positive cell fraction (p<0.002).

Figure 4.6. PG-rich lesions are associated the presence of Sox9-expressing cells(arrows). MP =

Movat’s pentachrome (blue = proteoglycan, yellow = collagen, black = nuclei/elastin, red =

cytoplasm/muscle). Scale bar = 100 µm.

MP

Sox9

Page 47: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

37

(A)

(B)

(C)

(D)

Figure 4.7. Relationship between Sox9 fraction and PG/GAG score in lesion areas. Error bars = SEM.

*p<0.005.

4.3.3 Lesions with dense morphology display distinct characteristics compared to those with diffuse morphology

Dense and diffuse lesion morphologies (Figure 4.1) were also characterized based on PG/GAG

content, ApoB lesion score, and lesion Sox9-positive cell fraction (Figure 4.8). Overall,

compared to lesions with a diffuse morphology, dense lesions expressed higher levels of

biglycan (p<0.001) and lower levels of hyaluronan (p=0.003). On average, dense lesions also had

higher ApoB scores (p=0.002) and a higher proportion of Sox9-expressing cells (p=0.006).

Dense lesions were more common in swine fed the HF/HC diet compared to normal chow at the

two-month time point (p=0.011). There was no difference in the frequency of dense and diffuse

lesions between swine fed the HF/HC diet compared to normal chow at the 5-month time point

and between swine from the 2- and 5-month time points fed the HF/HC diet.

*

*

*

Page 48: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

38

(A)

(B)

(C)

(D)

Figure 4.8. Dense and diffuse lesion characteristics and frequency with time points and diets.

Morphologically distinct lesions were compared based on (A) specific proteoglycan and

glycosaminoglycan (PG/GAG) content; (B) apolipoprotein B (ApoB) score; and (C) Sox9 fraction in

dense and diffuse lesions. (D) The number of dense lesions was compared between time points and diet

groups. Error bars = SEM. *p<0.05, **p<0.0125

*

*

*

*

**

Page 49: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

39

4.4 Discussion

The availability of suitable human aortic valve samples has limited the study of early CAVD.

The porcine model developed by Sider et al. [18, 19] successfully mimics several traits seen in

humans [11, 17] and allows for the study of the initial stages of disease pathogenesis, which

satisfies an unmet scientific need for a disease without an effective medical therapy. Valve

leaflets from this porcine model demonstrated the presence of lesions that accumulated PGs, a

feature that has been seen in calcified aortic valves [14] and early disease stages [16]. Here, the

quantification of PG/GAG content in the early CAVD lesions of these porcine valves, as well as

their association with lipid retention and putative chondrogenesis markers, was elucidated.

4.4.1 Biglycan may be involved in lipid retention and chondrogenesis in early lesions of CAVD

Notably, increases in biglycan with HF/HC diet were evident in lesions at the two-month time

point. Previously, biglycan has been observed in and surrounding small calcified nodules with

greater abundance than in and around larger calcified nodules [14]. Our observations reinforce

the supposition that biglycan is involved in early nodule formation. Often, in early nodules and

small calcified nodules from human aortic valves, biglycan is observed to co-localize with

decorin [14, 16]. At the two-month time point, decorin content also increased in lesions from

HF/HC diet-fed pigs, but this was not statistically significant. Within localized lesion areas,

increased biglycan score was also associated with increasing ApoB score and Sox9 fraction,

indicating that its role in early CAVD pathogenesis may involve lipid retention and early

chondrogenesis consistent with previous observations by Sider et al [19].

Biglycan has been observed to co-localize with apolipoproteins in human valves [16, 17] and

recently, was shown to mediate lipid retention in porcine aortic valves likely by binding LDL

lysine residues with its negatively charged GAG chains [50]. Certain PGs, particularly decorin,

have also been shown to act as accessory molecules to mediate LDL retention on collagen [50,

157]. While biglycan does bind to collagen in vitro [158], it is unknown whether it plays any role

in LDL-collagen interactions. In vitro studies show that biglycan is able to sequester TGF-β [14,

34], which in turn is able to stimulate VIC expression of PGs that have enhanced binding to LDL

[126]. The likely role of biglycan in LDL deposition as an initiating factor in CAVD is

unsurprising, as it would mimic findings from early human atherosclerosis [15].

Page 50: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

40

Lipid retention, whether directly or indirectly mediated by biglycan, allows modifications of

lipoproteins, which can perpetuate a cycle of additional lipid retention and inflammation. In

CAVD, biglycan induces the expression of phospholipid transfer protein (PLTP) by VICs

through interactions with Toll-like receptor 2 (TLR2) [129]. In atherosclerosis, PLTP promotes

modification of ApoAI, which increases its binding affinities for PGs [159] and may reduce the

ability of modified HDLs to perform reverse cholesterol transport [129, 159]. In human stenotic

valves, biglycan and ApoB also co-localize with oxidized LDL (oxLDL) [129], the accumulation

of which is strongly associated with increased inflammatory activity and tissue remodeling [51,

54]. Thus, the retention of lipoproteins by biglycan within valve lesions may promote enzyme-

mediated modification and oxidation by reactive oxygen intermediates or radicals generated by

cellular enzymes to promote the initiation of CAVD progression.

As well as an involvement in early lipid retention, biglycan may play a role in chondrogenic

processes in CAVD. Within lesion areas, increased Sox9 fraction was also associated with higher

biglycan scores. The transcription factor Sox9 is observed in both non-calcified pediatric and

calcified adult diseased valves [13]. Expression of Sox9, as well as other chondrogenic

transcription factors such as Twist1, Mef2c in putatively early and late-stage calcified valves,

may promote the production of a cartilage-like matrix that induces VIC activation in CAVD

pathogenesis [13]. Further, the soft microenvironment of these PG-rich lesions [18] may also

promote chondrogenic differentiation of VICs when stimulated by TGF-β [160], which can be

sequestered by biglycan [14, 34]. Soluble biglycan contributes to the pro-osteogenic effects of

oxLDL on VICs by increasing the expression of BMP2 and ALP through TLR2 [130]. Thus as

CAVD progresses, the role of biglycan in lesion formation may begin with chondrogenic

pathways that eventually lead to osteochondrogenic processes, resulting in the formation of

cartilage and bone that is often seen in stenotic valves [67, 86].

4.4.2 Hyaluronan may play a protective role in early CAVD lesion pathogenesis

In contrast to biglycan, hyaluronan content showed a decreasing trend with presumed lesion

severity. At two months, pigs fed the HF/HC diet demonstrated diminished hyaluronan content

when compared to pigs fed the normal chow diet. The decrease in hyaluronan with these more

advanced lesions suggests that the non-sulfated GAG plays a protective role during the early

stages of CAVD progression. In vitro, VICs interacting with hyaluronan through CD44

Page 51: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

41

demonstrate suppressed calcified nodule formation [21]. These results are recapitulated in vivo,

where the abundance of hyaluronan is inversely related to the magnitude of fibrosa calcification

[135]. Further, native porcine valve leaflets treated with anti-CD44 increase expression of ALP

and Runx2, all markers of VIC dysfunction [21]. At this early stage of disease progression

though, where calcification is not yet observed [19], hyaluronan may play a protective role

inhibiting lipid retention and putative chondrogenesis, since hyaluronan demonstrated an inverse

relationship with ApoB score and Sox9-positive cell fraction. Within the valve, hyaluronan

turnover and/or removal have been associated with hypoxia, osteogenesis, apoptosis, and cell

proliferation [21, 140], but the relationship of the GAG with lipid retention and chondrogenesis

has not been explored.

4.4.3 Early CAVD lesions demonstrate further ECM remodeling with distinct morphological characteristics

With pigs fed the HF/HC diet, levels of biglycan, decorin, and versican decreased between the

two- and five-month time points. Interestingly, although there were changes in specific PG/GAG

content, total PG content was not altered. This is explained in part by hyaluronan content being

unchanged within most lesion areas (Appendix A.3), with hyaluronan consistently comprising

~65% of the whole lesion proteoglycan area in lesions from pigs fed the HF/HC diet (Figs. 4.3

and 4.4), regardless of time on the diet. Further, while biglycan, decorin, and versican levels in

the HF/HC lesions decreased from two- to five-month time points, there was greater separation

(i.e., less co-localization) of areas positively stained for versican versus those positively stained

for biglycan and decorin in five-month HF/HC lesions compared to two-month HF/HC lesions.

This is consistent with reports from advanced stenotic lesions in which biglycan- and decorin-

stained areas co-localize but do not overlap with versican-stained areas [16]. In addition,

increases in collagen were observed between the two- and five-month HF/HC groups, suggesting

that as disease progresses there may be further remodeling of the ECM, where PGs are replaced

by other ECM components within the lesion. In contrast, previous studies of advanced CAVD

demonstrated decreased total collagen content within whole, calcified valves [79]. The

differences likely reflect different phases of remodeling (early fibrotic vs. late calcific), plus the

more localized and lesion-specific analysis in this study (vs. whole leaflet analysis).

Furthermore, comparisons of dense and diffuse lesions showed that morphological differences

may be useful in identifying early lesion stages. Compared to diffuse lesions, dense lesions had

Page 52: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

42

greater biglycan content and lower levels of hyaluronan. In these valve centre sections, dense

lesions had higher ApoB scores and increased proportions of Sox9-positive cells, which

recapitulates a previous study analyzing lesions in non-centre sections [18, 19]. These

observations indicate that lesions may progress from a more diffuse morphology to become

dense, accumulating biglycan, losing hyaluronan, and developing a lipid-retaining and putatively

chondrogenic profile in the process.

Dense lesions were found to be more common in swine fed the HF/HC diet compared to normal

chow diet after 2-months. Surprisingly, there was no propensity for dense lesion morphologies in

HF/HC diet swine compared to normal chow diet swine at 5-months, nor in 5-month swine

compared to 2-month swine fed the HF/HC diet, as might be expected if dense lesions

represented a more advanced morphology. Comparisons between swine fed for 2- and 5-months

on HF/HC diet may be affected by differences in when HF/HC diet was administered. Five-

month HF/HC swine began the atherogenic diet at an earlier age than two-month HF/HC swine.

This may confound temporal comparisons compared to if swine began HF/HC diet at the same

age with five-month swine receiving this diet for a longer period of time. It is also possible that

diffuse and dense lesions do not represent progressive stages of early lesion formation, but

entirely different early lesion types.

Interestingly, large dense PG-rich lesions occasionally appeared morphologically cartilaginous

with lipid near the surface and Sox9-expressing cells near the base of the lesion [18]. This

layered appearance is similar to the stratified appearance of macrophages in non-calcified and

early calcified lesions [11, 17] and provides further evidence of the active pathological processes

at work in early lesions. Proteoglycan, specifically biglycan, accumulation may promote the

retention of lipids and chondrogenesis, which eventually activate inflammatory pathways and

calcification.

This is the first instance when specific PGs/GAG were characterized within early CAVD lesions.

Previous studies of advanced CAVD lesions demonstrated the presence of biglycan and decorin

and absence of versican and hyaluronan immediately surrounding small calcified nodules [14].

This is consistent in part with observations in our early porcine model of CAVD, where increases

in biglycan and decreases in hyaluronan were observed within lesion areas. In contrast, previous

studies also showed that larger and possibly more advanced calcified nodules were surrounded

Page 53: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

43

by versican and hyaluronan, but not biglycan and decorin [14]. These characteristics of advanced

stenotic valves do not correlate with the observed PG/GAG changes observed in our model,

suggesting that there may be multiple shifts in ECM remodeling during CAVD progression. For

example, biglycan may increase in early CAVD lesions to promote lipid retention and eventual

early calcified nodule formation, but may be replaced by other components when calcification

becomes more severe. Furthermore, hyaluronan may play a protective role early on in CAVD

progression, as suggested by its absence in lesions in our early porcine model and around smaller

calcified nodules, but may play a role in continued mineralization of larger calcified nodules.

This also supported by changes observed in collagen content. In our early porcine model of

CAVD, we saw increases in total collagen content whereas in advanced stenotic valves, total

collagen content decreases [79]. While early lesions eventually lose PG and accumulate collagen,

in advanced lesions this collagen may become calcified. Overall, these observations are

suggestive of the continued ECM remodeling that occurs throughout CAVD progression.

4.5 Conclusion

The presence and role of PGs is only a recently studied component of CAVD. In a previous

study using this porcine model, the formation of PG-rich lesions in the absence of lipid

deposition, macrophages, osteoblasts, or myofibroblasts suggested an important role for this

ECM component in early CAVD stages. Through this histological study, possible roles of

specific PG/GAGs were elucidated. In particular, the accumulation of biglycan and loss of

hyaluronan within early lesions may play a role in lipid retention and putative chondrogenesis,

due to their association with ApoB and Sox9, respectively.

Page 54: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

44

Chapter 5

5 Phenotypes of valve interstitial cells in lesions of early calcific aortic valve disease

5.1 Introduction

Focal and layer-dependent susceptibility to calcific aortic valve disease (CAVD) suggests that

the valve is composed of a heterogeneous population of cells that are phenotypically different

based on their microenvironments. This has been determined in valvular endothelial cells

(VECs), which on the fibrosa side have enhanced anti-oxidative and calcification-permissive

characteristics compared to their ventricularis-side counterparts [39]. The spatial heterogeneity

of VECs also contributes to valvular interstitial cell (VIC) function, which is partly regulated by

VEC paracrine signaling [161-163]. Within the population of VICs, there is a subpopulation of

mesenchymal progenitor cells with adipogenic, chondrogenic, osteogenic, and myofibrogenic

potential [164]. Pathological cells, which are mostly myofibroblasts [41] and osteoblasts [67,

80], likely arise from these progenitors [20].

In addition to VEC paracrine signaling, VIC function is influenced by mechanical factors,

biochemical stimuli, and extracellular matrix (ECM) cues. VICs are shielded from the disturbed

hemodynamics on the fibrosa side and undisturbed shear stress conditions on the ventricularis

side, that likely affect VEC side-dependent pathosuceptibility. Instead, VICs experience

mechanical deformation through interactions with the ECM, which itself deforms as the valve

opens and closes. Pathological stretching of valve leaflets increases the expression of proteolytic

enzymes, pro-inflammatory proteins, and differentiation markers that are upregulated in diseased

states [165-167]. These changes are often seen in the fibrosa layer [165, 166], which is stiffer in

comparison to the ventricularis layer [168-170]. Within the individual valve layers, micropipette

aspiration indicates the presence of distinct soft and stiff regions [170]. Extracellular matrix

stiffness may directly affect VIC function, but also modulates VIC responses to biochemical

factors. For example, when grown in osteogenic differentiation media, VIC differentiation into

osteoblasts with the formation of bone nodules preferentially occurs on softer (~20 kPa)

substrates, while stiffer (>100 kPa) substrates promote differentiation into myofibroblasts [36,

Page 55: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

45

133]. Further, myofibroblast differentiation of VICs induced by transforming growth factor beta

1 (TGF-β1) only occurs on stiff substrates in a β-catenin dependent manner [37].

Myofibroblasts and osteoblasts are commonly observed in late stages of disease, but rarely in the

early stages. Changes in the valve microenvironment still occur, as ECM disorganization is a

major characteristic of both early and late stages of CAVD. Early valve disease is identified by

focal subendothelial thickening on the fibrosa side of the leaflet with accumulations of

proteoglycan, lipid, inflammatory cell infiltrate, and extracellular mineralization [11, 17]. Late-

stage valve disease continues with these ECM changes, but with greater severity, resulting in

fibrosis and calcification. Recently, a porcine model of early CAVD by Sider et al. [18, 19]

demonstrated the formation of proteoglycan (PG)-rich lesions on the pathosuceptible fibrosa side

of the valve. At this early stage of disease progression, myofibroblasts, osteoblasts, and

significant inflammatory infiltrate were absent, but lipid retention and putative chondrogenesis

were evidenced by the presence of ApoB and Sox9-expressing cells.

Micromechanical testing demonstrated that these PG-rich lesions are softer than adjacent non-

lesion fibrosa [18]. In addition, ApoB positive areas are often present immediately beneath the

subendothelium and Sox9-expressing cells are commonly observed at the base of these lesions.

These observations demonstrate that there are not only differences between lesion and non-lesion

areas, but spatially within lesions as well. Previous studies of native valve cell phenotype using

microarrays have only profiled VICs from the entire leaflet, effectively ignoring the putative

heterogeneity between lesion and non-lesion areas and within lesions [129, 171]. The focal

nature of valve disease and sensitivity of VICs to their changing microenvironment warrant the

study of VIC phenotype with a spatially directed approach. This chapter compares VIC

phenotypes in early CAVD lesion areas and non-lesion fibrosa.

5.2 Materials and Methods

5.2.1 Frozen valve leaflet section preparation

Left coronary valve leaflets from the early porcine model by Sider et al. [18, 19] were also used

for this chapter (see 4.2.1). Leaflets frozen in optimal cutting temperature (OCT) compound were

chosen for this objective, as they allow retrieval of higher quality RNA from laser capture

microdissection (LCM) than formalin-fixed, paraffin-embedded sections [172] (Appendix B.5).

Page 56: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

46

VICs were isolated and compared for differential gene expression in lesion and non-lesion areas.

Lesion areas included those (1) at the top half of the lesion, and (2) at the bottom half of the

lesion, where Sox9-expressing cells are often present [18]. Non-lesion areas were those far away

from lesions, but still within the fibrosa layer.

Figure 5.1. Cryosectioning slide schematic for each porcine sample. Serial sections for staining (MP =

Movat’s pentachrome; Sox9) and for RNA isolation (LCM = laser capture microdissection) allowed for

identification and tracking of lesion morphology during LCM. For each pig, four sets of slides (denoted

below the bracket) were prepared followed by two additional staining slides.

Eight-micrometer radial centre sections from the left coronary valve leaflets were cryosectioned

using the Leica CM 3050S cryostat in Dr. Philip Marsden’s Lab (Li Ka Shing Research Institute,

St. Michael’s Hospital, Toronto). Sections were mounted on charged slides to maximize

adherence of leaflet sections to slides during staining and of undesired tissue areas to slides

during LCM. For each pig sample, four sets of slides were prepared followed by two additional

staining slides. Each set contained serial sections on two staining slides and three LCM slides

(Figure 5.1). Each slide for staining had two tissue sections and each slide for LCM had four

tissue sections. Slides for LCM were kept at -80°C until use. Slides for staining were kept at

room temperature overnight to allow sections to better adhere and subsequently stored at -80°C

until staining.

5.2.2 Histological and immunohistochemical identification of lesions and samples of interest

According to Sider et al. [18, 19] and results from Chapter 4, lesions from two-month HF/HC

samples and dense lesions were most commonly associated with higher Sox9-positive cell

fraction. Therefore, dense lesions from two month HF/HC samples were used to probe VIC

phenotypic changes more broadly by microarray analysis. PG-rich lesions were identified on

sections stained with Movat’s pentachrome, using a modified protocol optimized for frozen

MP Sox9 MP Sox9 LCM LCM LCM

x4

Page 57: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

47

sections (Appendix B.6). Lesions were further narrowed down by Sox9 (anti-Sox9, rabbit

polyclonal, 2 µg/mL, ab3697, Abcam) immunohistochemical staining, which identified those

with putatively chondrogenic cells (Appendix B.7). Briefly, IHC began by warming the slides at

room temperature for 30 min. During this time, acetone was pre-cooled at -20°C for the

subsequent acetone fixation for 10 min at room temperature. Endogenous peroxidases were then

blocked with 3% H2O2/methanol for 10 min. Non-specific staining was blocked using 1% goat

serum buffer for 30 min prior to an hour-long incubation with primary antibody. Samples were

then incubated with biotin labeled anti-rabbit secondary (Vector Laboratories, Burlington, ON,

Canada) with 1.5% goat serum for 30 min. All samples were incubated with avidin-biotin-

peroxidase conjugate (Vectastain Elite ABC Kit, Vector Laboratories) for 30 min. Positive

staining was visualized following a 5 min incubation in Vector NovaRED (Vector Laboratories),

followed by Vector Hematoxylin QS (Vector Laboratories) counterstaining. PBS/Tween was

used to wash between steps. Negative controls involved no primary and IgG controls (R&D

Systems, Minneapolis, MN, USA). In total, four pig samples from the 2-month HF/HC group

were selected based on largest lesion size and presence of putatively chondrogenic cells, as

designated by Movat’s pentachrome and Sox9 staining, respectively (Figure 5.2).

5.2.3 Laser capture microdissection

Serial sections intended for LCM were quickly stained immediately prior to microdissection

using Arcturus Histogene Staining Solution (Life Technologies, Burlington, ON, Canada). All

three slides in each LCM set were stained and microdissected in one batch within an hour. For

each set, VICs were isolated with the Arcturus PixCell IIe (Li Ka Shing Research Institute) from

the areas of interest in the following order: (1) top of lesion; (2) bottom of lesion; and (3) non-

lesion areas. The smallest laser spot size, 7.5 µm, was used to ensure accurate capture of regions

of interest. The power of the pulse ranged from 60-80 mW and the duration of each pulse from

30.0-35.0 msec. The LCM environment was regulated by a dehumidifier, which maintained the

relative humidity below 35% in the room. For each area of interest, VICs were captured on

separate CapSure Macro LCM Caps (Life Technologies). For each pig, all nine microdissected

tissue samples were capture on the same day to reduce technical variability. Following 30 min

incubation in extraction buffer from the PicoPure RNA Isolation Kit (Life Technologies) at

42°C, samples were stored at -80°C until RNA isolation (Appendix B.8).

Page 58: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

48

(A)

(B)

(C)

(D)

Figure 5.2. Lesions of interest for differential gene expression analysis. The largest dense lesions (arrows)

from four pigs fed the HF/HC diet for two-months (labeled by numerical identifiers) were distinguished by

Movat’s pentachrome (MP) staining.

5.2.4 RNA isolation, amplification, and microarray analysis

RNA was isolated from all nine microdissected samples in a set at the same time using the

PicoPure RNA Isolation Kit (Life Technologies). DNase treatment was performed using a

RNase-free DNase Set (Qiagen, Gaithersburg, MD, USA). All samples were eluted with 11 µL

of elution buffer (Appendix B.9). After isolation, replicates of the areas of interest from each set

were pooled.

Samples were sent to the Ontario Cancer Institute Genomics Centre (Toronto, ON, Canada) for

downstream processing and statistical analysis. RNA quality was analyzed using a 2100

Bioanalyzer (Agilent Technologies Canada Inc., Mississauga, ON, Canada). All samples had

RNA integrity numbers between 2.3-7.0 and concentrations between 242-816 pg/µL (Appendix

A.5). RNA was amplified using the Ovation Pico Whole Transcript Amplification Kit (NuGen,

11107 11212

12509 11907

Page 59: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

49

San Carlos, CA, USA). The resulting cDNA was also run on the 2100 Bioanalyzer for quality

control (Appendix A.5). Profiles indicated suitable cDNA samples with curves that plateaued

around 200-500 nucleotides with some high molecular weight products around 500-1000

nucleotides. Amplified samples were run on the GeneChip 1.0ST Porcine Array (Affymetrix,

Santa Clara, CA, USA), where a total of 19 124 probe sets are represented.

5.2.5 Data processing and statistical analyses

Data was checked for overall quality using R (v2.15.3) with the Bioconductor framework and the

Array Quality Metrics package. After importing the data, a number of graphs were generated to

ascertain if there were any potential problems or outliers (Appendix A.6). A boxplot of

unprocessed log-intensity distributions and a histogram of the density of log-intensities

demonstrated similar probe intensities between arrays. The histogram of log-intensities was also

uni-modally distributed, which suggests the absence of artifacts that affect certain areas of the

arrays. This was recapitulated by 2D plots of expression characteristics (predicted by Probe

Level Model (PLM) estimates) at their array positions. Each array demonstrated homogeneous

colour coded values, implying that there was no spatial bias within the arrays. Relative Log

Expression (RLE) boxplots were constructed by calculating ratios between the expression of a

probe set and the median expression of this probe set across all arrays of the experiment. The

boxes were similar in range and centred close to 0, which is expected assuming only relatively

few genes are differentially expressed. Boxplots of Normalized Unscaled Standard Error (NUSE)

values indicated that array quality was satisfactory, as distributions centred around 1 (low quality

distributions centre around 1.1) and each array had a similar global spread. Finally, a correlation

coefficient for each pair of arrays was qualitatively presented on a coloured matrix. The minimal

coefficient value across all arrays was 0.90, indicating that there is homogeneity among array

intensities. Overall, all samples passed quality control and were included in subsequent analysis.

Data was imported into GeneSpring v12.6 for analysis. During import, the data was normalized

using a robust multiarray average (RMA) 16 normalization followed by a “per probe” median-

centred normalization, which are standard for Affymetrix ST arrays. All data analysis and

visualization were performed on log2 transformed data. To approximate the level of macrophage

contamination, non-median centred intensity averages for macrophage markers were categorized

as: (1) not expressed (<100); (2) lowly expressed (100-500); (3) moderately expressed (500-

Page 60: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

50

4000); and (4) highly expressed (>4000) (personal communication with Natalie Stickle, UHN

Microarray Centre). These values roughly correlated to the 25th

, 80th

, 95th

, and 99th

percentiles of

intensity levels, respectively. The average expression value for all transcripts was below the 75th

percentile of intensities.

For subsequent analyses, data were filtered to remove the confounding effect of probes that

showed no signal. Probes above the 20th

percentile of intensities from any of the groups of

interest were allowed to pass through this filtering. The final set contained 15 057 probe sets.

Repeated Measures ANOVA with a Benjamini-Hochberg false discovery rate (FDR) corrected

p<0.05 did not show any significant probe sets. Therefore, the repeated measures ANOVA was

repeated with an uncorrected p-value cut-off of 0.05 (Appendix A.7). The significant results of

this repeated measures ANOVA (1246 probesets) were analyzed by Tukey’s post-hoc tests. To

interpret these results, the fold change was calculated for each pair of interest and a cut-off of 1.5

fold up or down was applied. Benjamini and Yekutieli corrected (p<0.3) hypergeometric tests

were applied to look for enriched Gene Ontology (GO) categories that overlapped. Due to the

large number of entries and lack of complete porcine annotations, an exhaustive analysis of the

complete gene list is beyond the scope of this study. In some cases where GO terms were

missing, GO terms of homologous genes from Homo sapiens were used. Differentially expressed

genes with putative significance to CAVD pathology were identified and discussed with a focus

on enriched GO biological processes.

5.2.6 Venn diagram analysis

Venn diagram analysis was performed to determine genes that were modulated commonly in

different regions. Two-way Venn diagrams were created using the list of differentially expressed

entries from top vs. fibrosa and bottom vs. fibrosa Tukey’s post hoc tests and VENNY online

software (http://bioinfogp.cnb.csic.es/tools/venny/index.html, BioinfoGP Bioinformatics for

Genomics and Proteomics CNB-CSIC, Madrid, Spain) [173].

5.3 Results

5.3.1 Sample characterization

Valve interstitial cells (VICs) were isolated from the top of lesions, bottom of lesions, and

normal fibrosa of pigs fed the HF/HC diet for two months. Previously, the formation of PG-rich

Page 61: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

51

lesions at the two-month time point in this porcine model of CAVD were found to occur in the

absence of macrophages, as identified by immunohistochemical staining [18]. Markers of these

inflammatory cells expressed at a low level (roughly <500 intensity level) or were not expressed

(<100 intensity level), confirming the absence of these possible cell contaminants in the

differential gene expression analyses (Table 5.1).

Table 5.1. Expression levels of select macrophage-specific markers

Gene Name Top of Lesion Bottom of Lesion Normal Fibrosa

EMR1 76.2 84.1 136.1

CD14 118.2 111.7 133.4

LOC100520753 (CD68) 171.0 189.9 488.0

SIGLEC-1 (CD169) 187.5 208.2 196.4

ITGAM (Mac-1) 167.2 176.8 151.7

FCGR1A (CD64) 67.5 66.9 98.6

CD80 92.3 117.3 131.5

CD86 271.9 278.5 572.3

Average median centred values

Differential gene expression by cells from the (1) top half of early lesions, (2) bottom half of

early lesions, and (3) non-lesion fibrosa was assessed. Repeated Measures ANOVA found 1246

differentially expressed sequences (p<0.05). Tukey’s post-hoc tests (p<0.05) found that between

groups there were 525 differentially expressed probe sets between the top and bottom of lesions

(TvsB), 832 differentially expressed probe sets between the top of lesion and non-lesion areas

(TvsF), and 176 differentially expressed probe sets between bottom of lesion and non-lesion

areas (BvsF). When a fold change cutoff of 1.5 was applied to these post-hoc results, 156 TvsB,

215 TvsF, and 18 BvsF differentially expressed transcripts remained (Figure 5.3).

Page 62: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

52

(A)

(B)

(C)

Figure 5.3. Distribution of differentially expressed transcripts in lesion and non-lesion areas. (A) Top of lesion vs.

bottom of lesion; (B) Top of lesion vs. normal fibrosa; (C) Bottom of lesion vs. normal fibrosa.

5.3.2 Lesion and non-lesion VIC differential gene expression

Differentially expressed genes common between TvsF and BvsF comparisons identified

transcripts that were differentially expressed between the fibrosa and throughout the lesion. With

a fold change cutoff of 1.5, only 7 total entities were identified. Without fold change cutoffs,

TvsF and BvsF comparisons identified 82 transcripts, 39 upregulated and 43 downregulated, that

were differentially expressed in VICs throughout the lesion area compared to VICs from normal

fibrosa (Figure 5.4). Analysis of GO terms for transcripts differentially expressed throughout the

lesion found only a few enriched biological processes with putative relevance to valve disease,

such as immune response and regulation of apoptosis (Table 5.2).

(A)

(B)

Figure 5.4. Transcript expression in lesion areas. Blue regions represent transcripts differentially expressed

between top of lesion and normal fibrosa VICs (TvsF). Yellow regions represent transcripts differentially

expressed between bottom of lesion and normal fibrosa VICs (BvsF). Union regions represent transcripts that

are differentially expressed throughout the lesion vs. the fibrosa. [UP] = upregulated. [DOWN] =

downregulated. No fold change cut-off was applied.

Page 63: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

53

Table 5.2. Select differentially expressed genes between lesion (top and bottom) and non-lesion

areas

Gene Name L/F Regulation GO Biological Process

IL1R1 Up Cytokine-mediated signaling pathway

LOC100516004 Up Lipopolysaccharide biosynthetic process

IL16 Up Induction of positive chemotaxis

NFKB2 Up Toll-like receptor signaling pathwayǂ

SERBP1 Up Regulation of apoptotic processǂ

LAMTOR5 Up Negative regulation of apoptotic processǂ

EIF4E Up Cytokine-mediated signaling pathwayǂ

LIPG Down Lipid metabolic process

HBP15/L22 Down Alpha-beta T cell differentiation

RCAN2 Down Calcium-mediated signaling

SMAD6 Down Immune response, negative regulation of apoptotic

process, negative regulation of BMP signaling

pathway, transforming growth factor beta receptor

signaling pathway, cell substrate adhesion

IGF1R Down Immune response, positive/negative regulation of

MAPK cascade, negative regulation of apoptotic

process

AKAP6 Down cAMP-mediated signaling, cellular response to

cytokine stimulus

TTC8 Down Fat cell differentiation

CRY Down Oxidation-reduction process, fatty acid metabolic

process

No fold change cutoff was applied. L/F = regulation in whole lesion area compared to non-lesion fibrosa cells. GO =

Gene Ontology. IL1R1, interleukin 1 receptor type I; LOC100516004, cyclin-dependent kinases regulatory subunit

1-like; IL16, interleukin 16; NFKB2, nuclear factor of kappa light polypeptide gene enhancer in B-cells 2; SERBP1,

SERPINE1 mRNA binding protein 1; LAMTOR5, late endosomal/lysosomal adaptor, MAPK and MTOR activator

5; EIF4E, eukaryotic translation initiation factor 4E; LIPG, endothelial lipase; HBP15/L22, heparin-binding protein;

RCAN2, regulator of calcineurin 2; SMAD6, SMAD family member 6; IGF1R, insulin-like growth factor 1

receptor; AKAP6, A kinase (PRKA) anchor protein 6; TTC8, tetratricopeptide repeat domain 8; CRY, CRY protein.

ǂ Incomplete GO biological process terms from homologous Homo sapiens genes

5.3.3 Differential gene expression of VICs within lesion areas

Comparisons from the TvsB group were used to examine differences in VIC gene expression

within spatially distinct areas of the lesion. With a fold change cutoff of 1.5, there were 156

differentially expressed genes between the top and bottom of lesion areas. Benjamini Yekutieli

corrected (p<0.3) hypergeometric tests were used to look for enriched GO categories. Sequences

associated with a wide range of biological processes were altered in the different VIC

environments.

Page 64: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

54

Of the 35 downregulated transcripts, only 18 had GO biological process information. Genes

involved in mesenchymal cell development, mesenchymal cell differentiation, and cardiac

epithelial to mesenchymal transition, namely HEY1, ERBB4-like, and WNT16-like, were

enriched and downregulated in the top of lesions compared to the bottom of lesions. Of the 121

upregulated transcripts, 73 had GO biological process data. Upregulated GO biological processes

that are of putative relevance to early CAVD included those involved in lipid-related processes

(Table 5.3) and immune response (Table 5.4). The presence of increased lipid-related genes in

the top of lesions recapitulates the commonly observed presence of ApoB-positive areas in the

subendothelial regions of lesions (Chapter 4). In addition, several enriched GO molecular

function terms involved lipid-related processes, such as lipid binding, lipase activity, and

lipoprotein particle remodeling.

Table 5.3. Select lipid-related genes that are upregulated in the top of lesions

Gene Name T/B Fold Change T/F Fold Change GO biological process

CD36 5.84 4.92 LDL particle mediated signaling, lipid

localization, positive regulation of lipid storage

SMPDL3A 5.65 9.39 Lipid catabolic process

PLA2G7 4.65 5.10 LDL particle remodeling, lipid catabolic process

APOE 3.93 4.27 Lipid transport, LDL particle remodeling, lipid

catabolic process

MSR1 3.74 Lipoprotein transport, cholesterol transport,

positive regulation of cholesterol storage

LPL 3.03 2.99 Lipid catabolic process

LIPA 2.84 2.64 Lipid metabolic process

ABCA1 2.25 2.84 Lipid localization

IL18 1.96 2.22 Lipopolysaccharide mediated signaling pathway

FZD4 1.51 Lipid transport, lipid localization

T/B = top of lesion cells relative to bottom of lesion cells. T/F = top of lesion cells relative to non-lesion fibrosa

cells. GO = Gene Ontology. CD36, fatty acid translocase; SMPDL3A, sphingomyelin phosphodiesterase acid-like

3A; PLA2G7, phospholipase A2 group VII; APOE, apolipoprotein E; MSR1, macrophage scavenger receptor 1;

LPL, lipoprotein lipase; LIPA, lipase A; ABCA, ATP-binding cassette sub-family A member 1; IL18, interleukin

18; FZD4, frizzled family receptor 4.

Page 65: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

55

Table 5.4. Select immune-related genes that are upregulated in the top of lesions

Gene Name T/B Fold Change T/F Fold Change GO biological process

CXCL14 6.47 Immune response

CTSS 3.27 2.90 Adaptive immune response

GPR183 3.12 Immune response, leukocyte activation,

lymphocyte activation

CCL20 2.53 4.00 Immune response

CD83 2.13 Positive regulation of lymphocyte activation,

positive regulation of leukocyte activation,

positive regulation of T cell activation

IL8 2.04 1.67 Cell chemotaxis, Inflammatory response

IL18 1.96 2.22 Positive regulation of lymphocyte activation,

positive regulation of leukocyte activation,

positive regulation of T cell activation,

SLA-DQA1 1.80 Positive regulation of lymphocyte activation,

positive regulation of leukocyte activation,

positive regulation of T cell activation

TNFAIP8 1.77 1.44 Negative regulation of apoptotic process

ICAM1 1.71 1.74 Leukocyte cell-cell adhesion

SELP 1.62 1.59 Positive regulation of leukocyte migration

SLA-DQB1 1.61 Positive regulation of lymphocyte activation,

positive regulation of leukocyte activation,

positive regulation of T cell activation

VAV1 1.57 1.48 Leukocyte activation, lymphocyte activation

T/B = top of lesion cells relative to bottom of lesion cells. T/F = top of lesion cells relative to non-lesion fibrosa

cells. GO = Gene Ontology. CXCL14, chemokine (C-X-C motif) ligand 14; CTSS, cathepsin S; GPR183, G-protein

coupled receptor 183; CCL20, chemokine (C-C motif) ligand 20; CD83, B-cell activation protein; IL8, interleukin-

8; IL18, interleukin-18; SLA-DQA1, major histocompatibility antigen SLA-DQA; IL2RG, interleukin-2 receptor

gamma; TNFAIP8, tumor necrosis factor alpha-induced protein 8; SELP, selectin P; SLA-DQB1, major

histocompatibility antigen SLA-DQB1; VAV1, vav 1 guanine nucleotide exchange factor.

ECM remodeling is hallmark of both early and late stages of CAVD pathogenesis. Overall, GO

biological process analyses did not identify ECM remodeling as enriched. Using fold change

cutoffs and Tukey’s post hoc test results though, VIC expression of genes involved in ECM

disassembly and organization were found to be upregulated within the top of lesions compared to

the bottom of lesions (Table 5.5).

Further examination of differentially expressed transcripts found other pathways that may be

involved in early disease pathogenesis. In this early model of CAVD, Sox9-expressing cells are

commonly observed in the bottom of lesions, suggesting chondrogenesis may be involved in

CAVD progression of these areas. Although not found to be differentially expressed between

Page 66: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

56

valve areas, SOX9 was moderately to highly expressed in all samples analyzed (expression

values > 1200). Analysis of gene expression though showed upregulation of osteopontin in the

top of lesions compared to the bottom of lesions (T/B fold change =4.07), suggesting that

osteogenic processes may also be occurring at this early stage of disease.

Table 5.5. Select ECM remodeling-related genes that are upregulated in the top of lesions

Gene Name Gene Symbol T/B Fold Change T/F Fold Change

Cathepsin S CTSS 3.27 2.90

ADAM metallopeptidase domain 28 ADAM28 3.04 3.03

Cathepsin Z CTSZ 2.26 2.21

Matrix metalloproteinase 9 MMP9 1.96 1.81

Hyaluronidase 2 HYAL2 1.80 1.93

Matrix metalloproteinase 14 MMP14 1.64 1.77

Chondroitin sulfate synthase 1 CHSY1 1.52 2.04

Metalloproteinase inhibitor 1 TIMP1 1.52 1.67

T/B = top of lesion cells relative to bottom of lesion cells. T/F = top of lesion cells relative to non-lesion fibrosa

cells.

5.3.4 Differential gene expression of VICs from specific lesion areas and non-lesion areas

With a fold change cutoff of 1.5, 18 total transcripts were differentially expressed between the

bottom of lesions and non-lesion fibrosa. Of these transcripts, only eight were fully annotated.

Unsurprisingly, no GO categories were enriched from this comparison. Using the same fold

change criteria, 215 differentially expressed genes were identified between VICs from the top of

lesions and normal fibrosa. Many (42%) of the transcripts from the TvsF comparison were also

identified in TvsB (Figure 5.5). Notably, the majority of aforementioned genes upregulated and

with putative relevance to CAVD in the TvsB comparison are also upregulated in the TvsF

comparison (Tables 5.3, 5.4, 5.5). Similarly, enriched GO terms that are upregulated in TvsF

include those involved in lipid-related processes, immune and inflammatory response, and ECM

remodeling.

Page 67: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

57

(A)

(B)

Figure 5.4. Transcript expression in lesion areas. Blue regions represent transcripts differentially expressed

between top of lesion and normal fibrosa VICs (TvsF). Yellow regions represent transcripts differentially

expressed between top and bottom of lesion VICs (TvsB). Union regions represent transcripts that are

differentially expressed in the top of the lesion compared to other areas. [UP] = upregulated. [DOWN] =

downregulated.

Examining the top of lesions on an individual gene basis, there were other transcripts that were

differentially expressed within these areas. Oxidative stress seemed to play a greater role in

disease progression in these areas. Oxidized LDL (oxLDL) is able to bind scavenger receptors,

such as CD36 and lectin-type oxidized low density lipoprotein receptor 1 (LOX1), which were

upregulated in the top of lesions compared to other lesion areas (T/F fold change: 4.92 and 3.87,

respectively). As well, several genes relevant to osteochondrogenic pathways were upregulated

in the top of lesions (Table 5.6). Although toll-like receptor 4 (TLR4) is not categorized under an

osteochondrogenic biological process, stimulation of these receptors upregulates the expression

of osteogenic factors in VICs [174].

Table 5.6. Select differentially expressed genes that are upregulated in the top of lesions compared

to non-lesion areas.

Gene Symbol T/F Fold Change GO Biological Processes

SPP1 4.37 Biomineral tissue development, ossification

SULF1 2.43 Cartilage development, bone development, positive

regulation of BMP signaling pathway

CHSY1 2.04 Negative regulation of ossification

HYAL2 1.93 Cartilage development, hyaluronan catabolic process

TLR4 1.76 Inflammatory response, toll-like receptor signaling pathway

SLC20A1 1.52 Phosphate-containing compound metabolic process

T/B = top of lesion cells relative to non- lesion cells. T/F = top of lesion cells relative to non-lesion fibrosa cells. GO

= Gene Ontology. SPP1, osteopontin; SULF1, extracellular Sulf-1; CHSY1, chondroitin sulfate synthase 1; HYAL2,

hyaluronidase 2; TLR4, toll-like receptor 4; SLC20A1, solute carrier family 20 member 1. GO = Gene Ontology.

Page 68: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

58

5.4 Discussion

Studies of early CAVD using this porcine model demonstrate the accumulation of PG-rich

lesions on the fibrosa side of valve leaflets without the presence of significant myofibroblast,

osteoblast, and inflammatory cell infiltration, as assessed by immunohistochemical expression of

phenotypic markers. Differences in stiffness between lesion and non-lesion areas and distinct

regions of lipid retention and putative chondrogenesis within lesions indicate that there are

varying microenvironments within the valve leaflet. Here, we examined the spatial variability of

VIC phenotypes within lesion and non-lesion areas.

Firstly, we sought to distinguish lesion and non-lesion VIC phenotypes by finding similar

differentially expressed genes in the TvsF and BvsF comparisons. With a fold change cutoff of

1.5, few genes were identified within this category and consequently, did not identify enriched

GO terminologies. Without fold change cutoffs, 82 differentially expressed transcripts between

the whole lesion and non-lesion fibrosa were identified. These genes fell under broad GO

biological processes, but included some that signify possible immune response and apoptotic

processes occurring throughout the lesion. The small number of differentially expressed genes

between the whole lesion and non-lesion areas may be explained by heterogeneity within the

lesion or limitations of the LCM technology. Lesions from this porcine model of early CAVD

commonly demonstrated ApoB positive areas immediately below the subendothelium at the top

of lesions and Sox9-expressing cell at the base of lesions. This spatial variability within lesions

was reflected in the comparatively higher number of differentially expressed genes between the

top and bottom of lesion VICs.

In total, there were 156 differentially expressed genes between the top and bottom of lesion areas

when a fold change cut-off of 1.5 was applied. GO analysis of these genes identified

mesenchymal cell differentiation and proliferation genes, HEY1, ERBB4, and WNT16, as

enriched and downregulated in the top half of lesions. HEY1 and ERBB4 are involved in normal

valve development processes, which are thought to be involved in valve disease [175]. HEY1 is

a target gene in Notch signaling that plays a critical cooperative role with HEYL in epithelial to

mesenchymal transition of endocardial cells to promote endocardial cushion development [176].

Relevant to valve disease, downregulation of HEY1 transcription occurs with inhibition of Notch

signaling and is associated with activation of osteogenic markers and increased calcified nodule

Page 69: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

59

formation [150]. ERBB4 encodes an enzyme in the epidermal growth factor receptor (EGFR)

subfamily, which is required for semilunar valve development [177]. Expression of ERBB4 is

associated with left ventricular outflow defects, such as aortic valve stenosis, but its mechanism

in disease progress is not yet known [178]. Conditional knockout of NOTCH1 in mice has been

shown to alter the expression of neuregulin 1, a ligand of ERBB4, suggesting that there may be a

link between NOTCH and EGFR pathways [178, 179]. Although the non-canonical WNT16 has

not been studied in developing or diseased valves, it has been found to regulate the expression of

certain Notch ligands [180].

In the top of lesions, lipid-related, immune response, and ECM remodeling processes were also

enriched, but upregulated compared to bottom of lesion VICs. Many of these differentially

upregulated transcripts were also identified when comparing gene expression in the top of

lesions with non-lesion fibrosa areas. Overall, comparisons of the areas of interest demonstrated

that bottom of lesion VICs were more similar to non-lesion VICs than top of lesion VICs. With a

fold change cutoff of 1.5, only 18 transcripts were found to be differentially expressed in the

BvsF comparison. This raises the issue of the technical limitations of LCM in accurately

separating the top and bottom halves of lesion VICs. Although the smallest laser spot size was

used, it was difficult to capture lesion areas in precise halves while avoiding VECs. For all

samples, the top was captured first. In smaller lesions, this may have collected more than just

desired top half of the lesion, resulting in the capture of non-lesion VICs when attempting to

collect bottom of lesion VICs. This undoubtedly would affect the reliability of comparisons

involving bottom of lesion VICs. With this in mind, the remainder of discussion will focus on

differentially expressed genes between the top lesion and non-lesion areas.

In total, 162 genes were upregulated and 53 genes were downregulated in the top of lesions

compared to non-lesion fibrosa. Of relevance to CAVD, pathways involving lipid-, immune-,

and ECM remodeling-related genes were upregulated in the top of lesions, as well as transcripts

that are associated with osteochondrogenesis. When present, ApoB-positive areas were often

present near the top of lesions. In these areas, genes involved with lipid metabolism and

localization may promote early CAVD progression. Lipoprotein lipase (LPL) is thought to

sequester lipid to recruit macrophages, which in turn can produce more LPL and encourage

further lipid retention. The increase of LPL transcripts within lesion areas indicates that these

processes may be at work in early disease progression. Scavenger receptors, such as CD36 and

Page 70: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

60

LOX1, were also upregulated in the top of lesions and can increase the cellular uptake of

modified lipoproteins, namely oxLDL. The accumulation of oxLDL has been associated with

inflammation and calcification in valve disease [54, 56]. The increase of phospholipase A2 group

VII (PLA2G7) mRNA expression in these same areas provides a possible link between the

accumulation of oxLDL, inflammatory response, and mineralization. PLA2G7 encodes an

enzyme that can metabolize oxLDL into lysophosphatidylcholine (LPC), which is a powerful

inflammatory metabolite [181]. In isolated VICs, LPC elevates the expression of phosphate-

related proteins, including sodium-dependent phosphate cotransporter 1 (Pit1) and osteopontin

[182]. These are encoded by SLC20A1 and SPP1, respectively, and were also differentially

upregulated in top of lesion VICs. Within early lesions, Pit1 and osteopontin may induce

phosphate-mediated apoptosis and mineralization of VICs, as they have been shown to do in

vitro [183]. Other genes with relevance to osteochondrogenic processes include SULF1,

HYAL2, and CHSY1, which have roles in chondrocyte development and ossification.

In contrast to the immunostaining results, SOX9 mRNA expression was not found to be

differentially expressed between regions, either within a lesion or between a lesion and the

normal fibrosa. Instead, SOX9 expression was uniformly high in all areas, which is consistent

with observations in swine [18] and mice [184] that Sox9-positive cells are present throughout

the valve. The discrepancy between the IHC and microarray results with regards to differential

SOX9 expression between lesion and non-lesion areas may be due to differential regulation of

transcript vs. protein expression or that the microarray experiments had insufficient statistical

power to detect regional differences. The latter issue could be addressed by regional analysis of

SOX9 expression by qPCR, perhaps with additional samples to increase the sample size and

statistical power.

Previously, PG-rich lesions were also shown to form in the absence of macrophages and

dendritic cells [18]. These observations were reinforced by the examination of inflammatory cell

markers, which were not expressed or expressed lowly. Still, genes involved in chemotaxis and

positive regulation of inflammatory cell activation were differentially upregulated in the top of

lesions. Greater expression of toll-like receptor 4 (TLR4) has been associated with pro-

inflammatory and pro-osteogenic responses [185, 186]. For example, TLR4 has been shown to

stimulate expression of intracellular adhesion molecule (ICAM1) and osteopontin [186, 187].

Upregulation of both TLR4 and ICAM1 at the top of lesions indicate that interactions between

Page 71: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

61

these genes may promote leukocyte infiltration at this early stage of disease progression [186].

Interactions between the upregulated TLR4 and SPP1 genes also suggest pro-calcific pathways at

work. In minimally and heavily calcified stenotic valves, osteopontin is always present and

varies in proportion to calcium deposition [81]. Increased expression of osteopontin at the top of

lesions suggests that in early disease, it may act as a promoter of initial calcification.

Analysis of differentially expressed genes in the top of lesions also supports the notion of further

ECM remodeling. Increased expression of the elastolytic cathepsin S may be involved during the

initial stages in fragmenting and reduplicating the elastic lamina and/or during disease

progression in neoangiogenesis [188, 189]. In addition, upregulation of matrix

metalloproteinases (MMPs) -9 and -14, as well as metalloproteinase inhibitor 1 (TIMP1) may be

other sources of ECM remodeling, particularly changes in collagen, at this stage of lesion

development. Candidates responsible for the changes in PG/GAG content within lesions are also

indicated by the increased expression of hyaluronidase 2 (HYAL2) and chondroitin sulfate

synthase 1 (CHSY1). Interestingly, the differential expression of these genes corresponds with

the results from Chapter 4 where increases in biglycan and decreases in hyaluronan were

observed in lesions at this diet and time point.

5.5 Conclusion

Here, differential gene expression between lesion and non-lesion VICs was examined. Spatial

differences in VIC phenotypes between top of lesion and non-lesion areas were evident. In

particular, genes involved in lipid retention and metabolism, immune response, ECM

remodeling, and mineralization were identified as differentially upregulated in the top of lesion

VICs. While these findings do not demonstrate causality, they suggest correlative links between

valve microenvironment and VIC phenotype in early CAVD progression for further

investigation. Although histological studies demonstrate spatially different areas within lesions

with respect to lipid retention and putative chondrogenesis, few phenotypic differences were

observed between the top and bottom portions of the lesions, likely due to a combination of

small lesion size and limitations of LCM technology. Although a more detailed analysis of

microarray results was hampered by the lack of complete annotations for the porcine array, these

data nonetheless provide new insights into phenotypic changes in VICs and pathobiological

processes that occur early in valve disease.

Page 72: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

62

Chapter 6

6 Conclusions and Future Work

6.1 Conclusions

The prevalence and lack of effective medical treatments for CAVD expose an unmet scientific

need to improve our understanding of disease progression. Early CAVD changes are an untapped

area of study that would vastly improve our understanding of how the disease progresses to

result in calcified and stenotic valves and may uncover novel therapeutic targets. A porcine

model of early CAVD was developed by Sider et al., revealing the accumulation of PG-rich

lesions without myofibroblast, osteoblast, and inflammatory cell infiltration [18, 19]. In many

cases, these lesions were observed in the absence of ApoB staining, suggesting proteoglycan

accumulation occurs before lipid retention. Using histological and microarray analyses, we

sought to further characterize the lesion ECM with a focus on PG/GAG content, as well as the

VIC phenotypes contributing to and resulting from these changes in valve microenvironment.

In this thesis, putatively advanced early lesions were characterized as morphologically dense and

having greater biglycan, but less hyaluronan content. Chondroitin sulfate synthase 1 synthesizes

a GAG component of biglycan and hyaluronidase 2 degrades hyaluronan. Interestingly, in VICs

from the top of lesions their respective genes, CHSY1 and HYAL2, were upregulated compared

to those from non-lesion fibrosa. Although histological analyses did not reveal distinct changes

in total PG, collagen, and other (nuclei, cytoplasm, elastin) within lesions, upregulation of

mRNA of elastolytic proteases (cathepsins) and enzymes involved in collagen turnover (MMPs

and TIMPs) recapitulate that active ECM remodeling is occurring. This is also supported by the

time-dependent decrease in biglycan, decorin, and versican with atherogenic diet and contrasting

severity of morphologically diffuse and dense lesions.

In early lesion areas, increases in biglycan score and decreases in hyaluronan were associated

with higher ApoB scores and Sox9 fraction. Biglycan may play a role in early lipid retention and

putative chondrogenesis, while hyaluronan may have a more protective role. The accumulation

of lipid, whether directly or indirectly mediated by biglycan, may allow modification of

lipoproteins, which could initiate a cycle of further lipid retention and inflammation. Within

Page 73: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

63

lesion areas, several genes involved in lipid metabolic processes and lipid storage were

upregulated compared to non-lesion fibrosa areas. Interestingly, both biglycan and ApoB have

been shown to co-localize with oxLDL [129]. The upregulation of scavenger receptors within

lesion areas, suggest that cellular uptake of modified lipoproteins may occur and promote the

inflammatory response and calcification attributed to advanced stages of CAVD. PLA2G7,

SLC20A1, and SPP1 genes were also found to be upregulated within lesions and may connect

the oxidative transformation of accumulating lipids to mineralization. Furthermore, several genes

related to inflammatory and osteochondrogenic processes were differentially expressed between

top of lesion and non-lesion fibrosa.

The results from our histological and microarray analyses provide important insights into the

ECM and cellular changes that occur in early CAVD progression. Although our characterization

does not identify causative relationships, the correlational connections gained provide a stepping

stone for a better understanding of CAVD with the ultimate goal of finding an effective

therapeutic target.

6.2 Future Work

6.2.1 Further characterization of ECM changes in early CAVD lesions

Characterization of lesion collagen in relation to specific PG/GAG content would explore

another potential aspect of the role of PGs in disease progression. Biglycan and decorin are both

known to mediate collagen fibrillogenesis [14, 131], which could alter the biomechanical

properties of the lesion microenvironment. ROI analysis of Movat’s pentachrome collagen

staining with PG/GAG score could provide a general sense of collagen and specific PG/GAG

localization. Movat’s pentachrome sections could also be analyzed with an orientation-

independent birefringence imaging system (PolScope) to identify the amount of fibrillar collagen

present within lesion ROIs to determine its relationship with specific PGs/GAG.

To further complement the results from Chapter 4, IHC for chondroitin sulfate synthase 1 and

hyaluronidase 2 within lesions would allow for the examination of spatial, temporal, and diet-

related correlations with specific PG/GAGs. Spatial localization of these enzymes with the

chondroitin sulfate PGs biglycan and decorin, and hyaluronan would indicate potential players

responsible for the temporal and diet-related PG/GAG changes. Using Movat’s pentachrome

Page 74: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

64

staining, correlations between MMP/TIMPs with collagen content and the elastolytic cathepsin S

with elastin would provide further insight into the ECM remodeling processes at work.

6.2.2 Validation and pathway analysis of microarray results

Examination of select differentially expressed genes using quantitative polymerase chain

reaction (qPCR) would validate results from the microarray analyses. In addition, IHC staining

would validate these results at the protein level. Transcripts of interest for validation include the

ECM remodeling genes (CHYS1, HYAL2, CTSS, MMP9, TIMP1) and genes discussed above

that may connect lipid retention to more advanced CAVD processes (CD36, LOX1, PLA2G7,

SLC20A1, SPP1).

In order to take full advantage of the hypothesis-generating power of microarrays, gene

expression network analysis can be used to reveal the transcriptional regulatory pathways. This

would require full annotation of the porcine arrays based on BLAST to cross-reference porcine

sequences to the human genome. These annotations would allow us to use readily available

pathway analysis tools, such as Database for Annotation, Visualization and Integrated Discovery

(DAVID), to better understand the transcriptional pathways involved in early CAVD lesion

formation.

6.2.3 In vitro studies of biglycan influence on VIC function

Both IHC and microarray analyses provide correlational results from which further causative

hypotheses can be generated. Early CAVD lesions from this porcine model were softer than

normal fibrosa [18] and demonstrated spatial and temporal PG/GAG content changes. From

these results, we hypothesize soft lesions microenvironments that are rich in biglycan and

deficient in hyaluronan contribute to VIC osteochondrogenic differentiation. This may occur via

a direct matricellular effect in which the fate of adherent VICs is modulated by the mechanical

and biochemical properties of the early lesion ECM. To elucidate the effect of PG/GAG content

and lesion micromechanical environment, it is proposed that 3D polyethylene glycol (PEG)

hydrogels be used to mimic normal fibrosa tissue (elastic modulus 11 - 22 kPa; collagen

adhesion peptides) and early lesions (elastic modulus ~ 5 kPa; incorporating soluble biglycan).

Freshly isolated porcine VICs would be cultured within the hydrogels in DMEM with 10% fetal

bovine serum. To determine the effect of soluble biglycan on the expression of SOX9,

Page 75: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

65

SLC20A1, SPP1, RUNX2, and COL10A1, cells would be treated with recombinant biglycan

(0.05, 0.10, and 0.20 µg/mL) for 48 hours. Gene expression would be measured by qPCR, while

protein expression would be assayed using immunoblotting with quantification by gel

densitometry. Another non-exclusive mechanism by which early lesion PGs may contribute to

VIC pathological differentiation is through their retention of LDL and presentation of oxidized

LDL to VICs. This hypothesized mechanism is supported in part by two recent studies [126,

190], but these studies used standard tissue culture plates that do not mimic the soft and distinct

mechanical environments in the lesion vs. fibrosa. Thus, the effect of oxLDL on VIC expression

of osteochondral genes and proteins could be tested in the hydrogel models proposed above.

6.2.4 Mechanistic studies of hyaluronan interaction with VICs

In our study, hyaluronan was suggested to have a protective role in early CAVD, as it was

associated with areas that had lower ApoB scores and Sox9 fractions. Similarly, in a study of

advanced, calcified aortic valves, hyaluronan turnover was associated with markers of hypoxia

and ossification [140]. Positive expression of HA binding receptors RHAMM and HARE are

observed surrounding calcified nodules, while CD44 is not strongly correlated with calcification.

Hyaluronan binding to each of these three receptors has been associated with the activation and

propagation of signaling pathways, such as ERK 1 and 2 [191, 192]. Interestingly, it has been

established that hyaluronan plays different roles depending on its molecular weight [193-195]. In

vitro treatment of VICs with different molecular weights of soluble hyaluronan coupled with

inhibition of RHAMM, HARE, and CD44 receptors could help to elucidate the mechanisms by

which hyaluronan mediates its protective role in early CAVD lesions. Specifically, this would be

achieved by monitoring calcified nodule formation and quantification of markers involved in

VIC dysfunction, such as αSMA, by Western blotting.

Page 76: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

66

References

[1] N. M. Rajamannan, R. O. Bonow, and S. H. Rahimtoola, "Calcific aortic stenosis: an

update," Nat Clin Pract Cardiovasc Med, vol. 4, pp. 254-62, May 2007.

[2] A. S. Go, D. Mozaffarian, V. L. Roger, E. J. Benjamin, J. D. Berry, W. B. Borden, D. M.

Bravata, S. Dai, E. S. Ford, C. S. Fox, S. Franco, H. J. Fullerton, C. Gillespie, S. M.

Hailpern, J. A. Heit, V. J. Howard, M. D. Huffman, B. M. Kissela, S. J. Kittner, D. T.

Lackland, J. H. Lichtman, L. D. Lisabeth, D. Magid, G. M. Marcus, A. Marelli, D. B.

Matchar, D. K. McGuire, E. R. Mohler, 3rd, C. S. Moy, M. E. Mussolino, G. Nichol, N.

P. Paynter, P. J. Schreiner, P. D. Sorlie, J. Stein, T. N. Turan, S. S. Virani, N. D. Wong,

D. Woo, and M. B. Turner, "Heart disease and stroke statistics--2013 update: a report

from the American Heart Association," Circulation, vol. 127, pp. e6-e245, Jan 1 2013.

[3] B. F. Stewart, D. Siscovick, B. K. Lind, J. M. Gardin, J. S. Gottdiener, V. E. Smith, D.

W. Kitzman, and C. M. Otto, "Clinical factors associated with calcific aortic valve

disease. Cardiovascular Health Study," J Am Coll Cardiol, vol. 29, pp. 630-4, Mar 1

1997.

[4] C. M. Otto, B. K. Lind, D. W. Kitzman, B. J. Gersh, and D. S. Siscovick, "Association of

aortic-valve sclerosis with cardiovascular mortality and morbidity in the elderly," N Engl

J Med, vol. 341, pp. 142-7, Jul 15 1999.

[5] G. M. Novaro, R. Katz, R. J. Aviles, J. S. Gottdiener, M. Cushman, B. M. Psaty, C. M.

Otto, and B. P. Griffin, "Clinical factors, but not C-reactive protein, predict progression

of calcific aortic-valve disease: the Cardiovascular Health Study," J Am Coll Cardiol,

vol. 50, pp. 1992-8, Nov 13 2007.

[6] A. Starr, C. L. Fessler, G. Grunkemeier, and G. W. He, "Heart valve replacement

surgery: past, present and future," Clin Exp Pharmacol Physiol, vol. 29, pp. 735-8, Aug

2002.

[7] S. J. Cowell, D. E. Newby, R. J. Prescott, P. Bloomfield, J. Reid, D. B. Northridge, and

N. A. Boon, "A randomized trial of intensive lipid-lowering therapy in calcific aortic

stenosis," N Engl J Med, vol. 352, pp. 2389-97, Jun 9 2005.

[8] A. B. Rossebo, T. R. Pedersen, K. Boman, P. Brudi, J. B. Chambers, K. Egstrup, E.

Gerdts, C. Gohlke-Barwolf, I. Holme, Y. A. Kesaniemi, W. Malbecq, C. A. Nienaber, S.

Ray, T. Skjaerpe, K. Wachtell, R. Willenheimer, and S. Investigators, "Intensive lipid

lowering with simvastatin and ezetimibe in aortic stenosis," N Engl J Med, vol. 359, pp.

1343-56, Sep 25 2008.

[9] K. L. Chan, K. Teo, J. G. Dumesnil, A. Ni, and J. Tam, "Effect of Lipid lowering with

rosuvastatin on progression of aortic stenosis: results of the aortic stenosis progression

observation: measuring effects of rosuvastatin (ASTRONOMER) trial," Circulation, vol.

121, pp. 306-14, Jan 19 2010.

Page 77: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

67

[10] K. D. O'Brien, "Pathogenesis of calcific aortic valve disease: a disease process comes of

age (and a good deal more)," Arterioscler Thromb Vasc Biol, vol. 26, pp. 1721-8, Aug

2006.

[11] C. M. Otto, J. Kuusisto, D. D. Reichenbach, A. M. Gown, and K. D. O'Brien,

"Characterization of the early lesion of 'degenerative' valvular aortic stenosis.

Histological and immunohistochemical studies," Circulation, vol. 90, pp. 844-53, Aug

1994.

[12] C. M. Otto, "Calcific aortic valve disease: new concepts," Semin Thorac Cardiovasc

Surg, vol. 22, pp. 276-84, Winter 2010.

[13] E. E. Wirrig, R. B. Hinton, and K. E. Yutzey, "Differential expression of cartilage and

bone-related proteins in pediatric and adult diseased aortic valves," J Mol Cell Cardiol,

vol. 50, pp. 561-9, Mar 2011.

[14] E. H. Stephens, J. G. Saltarrelli, L. S. Baggett, I. Nandi, J. J. Kuo, A. R. Davis, E. A.

Olmsted-Davis, M. J. Reardon, J. D. Morrisett, and K. J. Grande-Allen, "Differential

proteoglycan and hyaluronan distribution in calcified aortic valves," Cardiovasc Pathol,

vol. 20, pp. 334-42, Nov-Dec 2011.

[15] Y. Nakashima, H. Fujii, S. Sumiyoshi, T. N. Wight, and K. Sueishi, "Early human

atherosclerosis: accumulation of lipid and proteoglycans in intimal thickenings followed

by macrophage infiltration," Arterioscler Thromb Vasc Biol, vol. 27, pp. 1159-65, May

2007.

[16] K. D. O'Brien, C. M. Otto, D. D. Reichenbach, C. E. Alpers, and T. N. Wight, "Regional

accumulation of proteoglycans in lesions of "degenerative" valvular aortic stenosis and

their relationship to apolipoproteins," 1995, p. 612.

[17] K. D. O'Brien, D. D. Reichenbach, S. M. Marcovina, J. Kuusisto, C. E. Alpers, and C. M.

Otto, "Apolipoproteins B, (a), and E accumulate in the morphologically early lesion of

'degenerative' valvular aortic stenosis," Arterioscler Thromb Vasc Biol, vol. 16, pp. 523-

32, Apr 1996.

[18] K. L. Sider, "Mechanical and Histological Characterization of Porcine Aortic Valves

under Normal and Hypercholesterolemic Conditions," Ph.D., Biomedical Engineering,

University of Toronto, Canada, 2013.

[19] K. L. Sider, C. Zhu, A. V. Kwong, Z. Mirzaei, C. F. M. de Lange, and C. A. Simmons,

"Evaluation of a porcine model of early aortic valve sclerosis," Cardiovascular

Pathology, 2014, In press.

[20] C. Y. Yip and C. A. Simmons, "The aortic valve microenvironment and its role in calcific

aortic valve disease," Cardiovasc Pathol, vol. 20, pp. 177-82, May-Jun 2011.

[21] K. J. Rodriguez, L. M. Piechura, and K. S. Masters, "Regulation of valvular interstitial

cell phenotype and function by hyaluronic acid in 2-D and 3-D culture environments,"

Matrix Biol, vol. 30, pp. 70-82, Jan 2011.

Page 78: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

68

[22] F. J. Schoen, "Evolving concepts of cardiac valve dynamics: the continuum of

development, functional structure, pathobiology, and tissue engineering," Circulation,

vol. 118, pp. 1864-80, Oct 28 2008.

[23] E. Rabkin-Aikawa, M. Farber, M. Aikawa, and F. J. Schoen, "Dynamic and reversible

changes of interstitial cell phenotype during remodeling of cardiac valves," J Heart Valve

Dis, vol. 13, pp. 841-7, Sep 2004.

[24] M. Misfeld and H. H. Sievers, "Heart valve macro- and microstructure," Philos Trans R

Soc Lond B Biol Sci, vol. 362, pp. 1421-36, Aug 29 2007.

[25] R. B. Hinton and K. E. Yutzey, "Heart valve structure and function in development and

disease," Annu Rev Physiol, vol. 73, pp. 29-46, 2011.

[26] E. H. Stephens, C. K. Chu, and K. J. Grande-Allen, "Valve proteoglycan content and

glycosaminoglycan fine structure are unique to microstructure, mechanical load and age:

Relevance to an age-specific tissue-engineered heart valve," Acta Biomater, vol. 4, pp.

1148-60, Sep 2008.

[27] M. J. Scott and I. Vesely, "Morphology of porcine aortic valve cusp elastin," J Heart

Valve Dis, vol. 5, pp. 464-71, Sep 1996.

[28] N. Latif, P. Sarathchandra, P. M. Taylor, J. Antoniw, and M. H. Yacoub, "Localization

and pattern of expression of extracellular matrix components in human heart valves," J

Heart Valve Dis, vol. 14, pp. 218-27, Mar 2005.

[29] K. J. Grande-Allen, N. Osman, M. L. Ballinger, H. Dadlani, S. Marasco, and P. J. Little,

"Glycosaminoglycan synthesis and structure as targets for the prevention of calcific aortic

valve disease," Cardiovasc Res, vol. 76, pp. 19-28, Oct 1 2007.

[30] J. R. Couchman and C. A. Pataki, "An introduction to proteoglycans and their

localization," J Histochem Cytochem, vol. 60, pp. 885-97, Dec 2012.

[31] L. Li, M. Ly, and R. J. Linhardt, "Proteoglycan sequence," Mol Biosyst, vol. 8, pp. 1613-

25, Jun 2012.

[32] S. Torii, R. I. Bashey, and K. Nakao, "Acid mucopolysaccharide composition of human-

heart valve," Biochim Biophys Acta, vol. 101, pp. 285-91, Nov 1 1965.

[33] M. M. Baig, G. R. Daicoff, and E. M. Ayoub, "Comparative studies of the acid

mucopolysaccharide composition of rheumatic and normal heart valves in man," Circ

Res, vol. 42, pp. 271-5, Feb 1978.

[34] H. Kresse, H. Hausser, E. Schonherr, and K. Bittner, "Biosynthesis and interactions of

small chondroitin/dermatan sulphate proteoglycans," Eur J Clin Chem Clin Biochem, vol.

32, pp. 259-64, Apr 1994.

Page 79: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

69

[35] J. H. Chen and C. A. Simmons, "Cell-matrix interactions in the pathobiology of calcific

aortic valve disease: critical roles for matricellular, matricrine, and matrix mechanics

cues," Circ Res, vol. 108, pp. 1510-24, Jun 10 2011.

[36] C. Y. Yip, J. H. Chen, R. Zhao, and C. A. Simmons, "Calcification by valve interstitial

cells is regulated by the stiffness of the extracellular matrix," Arterioscler Thromb Vasc

Biol, vol. 29, pp. 936-42, Jun 2009.

[37] J. H. Chen, W. L. Chen, K. L. Sider, C. Y. Yip, and C. A. Simmons, "beta-catenin

mediates mechanically regulated, transforming growth factor-beta1-induced

myofibroblast differentiation of aortic valve interstitial cells," Arterioscler Thromb Vasc

Biol, vol. 31, pp. 590-7, Mar 2011.

[38] S. T. Gould, S. Srigunapalan, C. A. Simmons, and K. S. Anseth, "Hemodynamic and

Cellular Response Feedback in Calcific Aortic Valve Disease," Circulation Research,

vol. 113, pp. 186-197, Jul 2013.

[39] C. A. Simmons, G. R. Grant, E. Manduchi, and P. F. Davies, "Spatial heterogeneity of

endothelial phenotypes correlates with side-specific vulnerability to calcification in

normal porcine aortic valves," Circ Res, vol. 96, pp. 792-9, Apr 15 2005.

[40] A. Bairati and S. DeBiasi, "Presence of a smooth muscle system in aortic valve leaflets,"

Anat Embryol (Berl), vol. 161, pp. 329-40, 1981.

[41] P. M. Taylor, S. P. Allen, and M. H. Yacoub, "Phenotypic and functional characterization

of interstitial cells from human heart valves, pericardium and skin," J Heart Valve Dis,

vol. 9, pp. 150-8, Jan 2000.

[42] M. Cimini, K. A. Rogers, and D. R. Boughner, "Smoothelin-positive cells in human and

porcine semilunar valves," Histochem Cell Biol, vol. 120, pp. 307-17, Oct 2003.

[43] S. A. Dreger, P. Thomas, E. Sachlos, A. H. Chester, J. T. Czernuszka, P. M. Taylor, and

M. H. Yacoub, "Potential for synthesis and degradation of extracellular matrix proteins

by valve interstitial cells seeded onto collagen scaffolds," Tissue Eng, vol. 12, pp. 2533-

40, Sep 2006.

[44] S. Sell and R. E. Scully, "Aging changes in the aortic and mitral valves: histologic and

histochemical studies, with observations on the pathogenesis of calcific aortic stenosis

and calcification of the mitral annulus," Am J Pathol, vol. 46, pp. 345-65, Mar 1965.

[45] N. M. Rajamannan, "Calcific aortic stenosis: lessons learned from experimental and

clinical studies," Arterioscler Thromb Vasc Biol, vol. 29, pp. 162-8, Feb 2009.

[46] R. V. Freeman and C. M. Otto, "Spectrum of calcific aortic valve disease: pathogenesis,

disease progression, and treatment strategies," Circulation, vol. 111, pp. 3316-26, Jun 21

2005.

Page 80: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

70

[47] M. A. Nicosia, R. P. Cochran, D. R. Einstein, C. J. Rutland, and K. S. Kunzelman, "A

coupled fluid-structure finite element model of the aortic valve and root," J Heart Valve

Dis, vol. 12, pp. 781-9, Nov 2003.

[48] J. J. Chiu and S. Chien, "Effects of disturbed flow on vascular endothelium:

pathophysiological basis and clinical perspectives," Physiol Rev, vol. 91, pp. 327-87, Jan

2011.

[49] M. A. Guerraty, G. R. Grant, J. W. Karanian, O. A. Chiesa, W. F. Pritchard, and P. F.

Davies, "Hypercholesterolemia induces side-specific phenotypic changes and peroxisome

proliferator-activated receptor-gamma pathway activation in swine aortic valve

endothelium," Arterioscler Thromb Vasc Biol, vol. 30, pp. 225-31, Feb 2010.

[50] E. B. Neufeld, L. M. Zadrozny, D. Phillips, A. Aponte, Z. X. Yu, and R. S. Balaban,

"Decorin and biglycan retain LDL in disease-prone valvular and aortic subendothelial

intimal matrix," Atherosclerosis, vol. 233, pp. 113-21, Mar 2014.

[51] M. Olsson, J. Thyberg, and J. Nilsson, "Presence of oxidized low density lipoprotein in

nonrheumatic stenotic aortic valves," Arterioscler Thromb Vasc Biol, vol. 19, pp. 1218-

22, May 1999.

[52] K. W. Walton, N. Williamson, and A. G. Johnson, "The pathogenesis of atherosclerosis

of the mitral and aortic valves," J Pathol, vol. 101, pp. 205-20, Jul 1970.

[53] J. Kuusisto, K. Rasanen, T. Sarkioja, E. Alarakkola, and V. M. Kosma, "Atherosclerosis-

like lesions of the aortic valve are common in adults of all ages: a necropsy study,"

Heart, vol. 91, pp. 576-82, May 2005.

[54] D. Mohty, P. Pibarot, J. P. Despres, C. Cote, B. Arsenault, A. Cartier, P. Cosnay, C.

Couture, and P. Mathieu, "Association between plasma LDL particle size, valvular

accumulation of oxidized LDL, and inflammation in patients with aortic stenosis,"

Arterioscler Thromb Vasc Biol, vol. 28, pp. 187-93, Jan 2008.

[55] A. Mahmut, M. C. Boulanger, D. Fournier, C. Couture, S. Trahan, S. Page, B. Arsenault,

J. P. Despres, P. Pibarot, and P. Mathieu, "Lipoprotein lipase in aortic valve stenosis is

associated with lipid retention and remodelling," Eur J Clin Invest, vol. 43, pp. 570-8,

Jun 2013.

[56] E. R. Mohler, 3rd, M. K. Chawla, A. W. Chang, N. Vyavahare, R. J. Levy, L. Graham,

and F. H. Gannon, "Identification and characterization of calcifying valve cells from

human and canine aortic valves," J Heart Valve Dis, vol. 8, pp. 254-60, May 1999.

[57] N. A. Nadlonek, J. H. Lee, M. J. Weyant, X. Meng, and D. A. Fullerton, "ox-LDL

induces PiT-1 expression in human aortic valve interstitial cells," J Surg Res, vol. 184,

pp. 6-9, Sep 2013.

[58] M. Olsson, C.-J. Dalsgaard, A. Haegerstrand, M. r. Rosenqvist, L. Rydén, and J.

Nilsson, "Accumulation of T lymphocytes and expression of interleukin-2 receptors in

Page 81: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

71

nonrheumatic stenotic aortic valves," Journal of the American College of Cardiology,

vol. 23, pp. 1162-1170, 1994.

[59] M. Olsson, M. Rosenqvist, and J. Nilsson, "Expression of HLA-DR antigen and smooth

muscle cell differentiation markers by valvular fibroblasts in degenerative aortic

stenosis," J Am Coll Cardiol, vol. 24, pp. 1664-71, Dec 1994.

[60] B. A. Warren and J. L. Yong, "Calcification of the aortic valve: its progression and

grading," Pathology, vol. 29, pp. 360-8, Nov 1997.

[61] M. E. Edep, J. Shirani, P. Wolf, and D. L. Brown, "Matrix metalloproteinase expression

in nonrheumatic aortic stenosis," Cardiovascular Pathology, vol. 9, pp. 281-286, Sep-Oct

2000.

[62] S. Helske, K. A. Lindstedt, M. Laine, M. Mayranpaa, K. Werkkala, J. Lommi, H. Turto,

M. Kupari, and P. T. Kovanen, "Induction of local angiotensin II-producing systems in

stenotic aortic valves," J Am Coll Cardiol, vol. 44, pp. 1859-66, Nov 2 2004.

[63] J. J. Kaden, C. E. Dempfle, R. Grobholz, C. S. Fischer, D. C. Vocke, R. Kilic, A. Sarikoc,

R. Pinol, S. Hagl, S. Lang, M. Brueckmann, and M. Borggrefe, "Inflammatory regulation

of extracellular matrix remodeling in calcific aortic valve stenosis," Cardiovasc Pathol,

vol. 14, pp. 80-7, Mar-Apr 2005.

[64] N. Cote, A. Mahmut, Y. Bosse, C. Couture, S. Page, S. Trahan, M. C. Boulanger, D.

Fournier, P. Pibarot, and P. Mathieu, "Inflammation is associated with the remodeling of

calcific aortic valve disease," Inflammation, vol. 36, pp. 573-81, Jun 2012.

[65] L. Wallby, T. Steffensen, and M. Broqvist, "Role of inflammation in nonrheumatic,

regurgitant heart valve disease. A comparative, descriptive study regarding

apolipoproteins and inflammatory cells in nonrheumatic heart valve disease," Cardiovasc

Pathol, vol. 16, pp. 171-8, May-Jun 2007.

[66] I. Perrotta, E. Russo, C. Camastra, G. Filice, G. Di Mizio, F. Colosimo, P. Ricci, S.

Tripepi, A. Amorosi, F. Triumbari, and G. Donato, "New evidence for a critical role of

elastin in calcification of native heart valves: immunohistochemical and ultrastructural

study with literature review," Histopathology, vol. 59, pp. 504-513, Sep 2011.

[67] E. R. Mohler, 3rd, F. Gannon, C. Reynolds, R. Zimmerman, M. G. Keane, and F. S.

Kaplan, "Bone formation and inflammation in cardiac valves," Circulation, vol. 103, pp.

1522-8, Mar 20 2001.

[68] E. Wypasek, J. Natorska, G. Grudzien, G. Filip, J. Sadowski, and A. Undas, "Mast cells

in human stenotic aortic valves are associated with the severity of stenosis,"

Inflammation, vol. 36, pp. 449-56, Apr 2013.

[69] O. Fondard, D. Detaint, B. Lung, C. Choqueux, H. Adle-Biassette, M. Jarraya, U. Hvass,

J.-P. Couetil, D. Henin, J.-B. Michel, A. Vahanian, and M.-P. Jacob, "Extracellular

matrix remodelling in human aortic valve disease: the role of matrix metalloproteinases

and their tissue inhibitors," European Heart Journal, vol. 26, pp. 1333-1341, Jul 2005.

Page 82: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

72

[70] B. Jian, P. L. Jones, Q. Y. Li, E. R. Mohler, 3rd, F. J. Schoen, and R. J. Levy, "Matrix

metalloproteinase-2 is associated with tenascin-C in calcific aortic stenosis," American

Journal of Pathology, vol. 159, pp. 321-327, Jul 2001.

[71] S. Helske, R. Oksjoki, K. A. Lindstedt, J. Lommi, H. Turto, K. Werkkala, M. Kupari, and

P. T. Kovanen, "Complement system is activated in stenotic aortic valves,"

Atherosclerosis, vol. 196, pp. 190-200, Jan 2008.

[72] M. ter Weeme, A. B. Vonk, K. Kupreishvili, M. van Ham, S. Zeerleder, D. Wouters, W.

Stooker, L. Eijsman, V. W. Van Hinsbergh, P. A. Krijnen, and H. W. Niessen, "Activated

complement is more extensively present in diseased aortic valves than naturally occurring

complement inhibitors: a sign of ongoing inflammation," Eur J Clin Invest, vol. 40, pp. 4-

10, Jan 2010.

[73] J. D. Miller, Y. Chu, R. M. Brooks, W. E. Richenbacher, R. Pena-Silva, and D. D.

Heistad, "Dysregulation of antioxidant mechanisms contributes to increased oxidative

stress in calcific aortic valvular stenosis in humans," Journal of the American College of

Cardiology, vol. 52, pp. 843-850, Sep 2008.

[74] K. Akat, M. Borggrefe, and J. J. Kaden, "Aortic valve calcification: basic science to

clinical practice," Heart, vol. 95, pp. 616-23, Apr 2009.

[75] K. D. O'Brien, D. M. Shavelle, M. T. Caulfield, T. O. McDonald, K. Olin-Lewis, C. M.

Otto, and J. L. Probstfield, "Association of angiotensin-converting enzyme with low-

density lipoprotein in aortic valvular lesions and in human plasma," Circulation, vol. 106,

pp. 2224-30, Oct 22 2002.

[76] N. K. Ghaisas, J. B. Foley, D. S. O'Briain, P. Crean, D. Kelleher, and M. Walsh,

"Adhesion molecules in nonrheumatic aortic valve disease: endothelial expression, serum

levels and effects of valve replacement," J Am Coll Cardiol, vol. 36, pp. 2257-62, Dec

2000.

[77] A. Mazzone, M. C. Epistolato, R. De Caterina, S. Storti, S. Vittorini, S. Sbrana, J.

Gianetti, S. Bevilacqua, M. Glauber, A. Biagini, and P. Tanganelli, "Neoangiogenesis, T-

Lymphocyte infiltration, and heat shock protein-60 are biological hallmarks of an

immunomediated inflammatory process in end-stage calcified aortic valve stenosis,"

Journal of the American College of Cardiology, vol. 43, pp. 1670-1676, May 5 2004.

[78] S. Chakraborty, E. E. Wirrig, R. B. Hinton, W. H. Merrill, D. B. Spicer, and K. E.

Yutzey, "Twist1 promotes heart valve cell proliferation and extracellular matrix gene

expression during development in vivo and is expressed in human diseased aortic

valves," Developmental Biology, vol. 347, pp. 167-179, Nov 1 2010.

[79] H. A. Eriksen, J. Satta, J. Risteli, M. Veijola, P. Vare, and Y. Soini, "Type I and type III

collagen synthesis and composition in the valve matrix in aortic valve stenosis,"

Atherosclerosis, vol. 189, pp. 91-8, Nov 2006.

[80] N. M. Rajamannan, M. Subramaniam, D. Rickard, S. R. Stock, J. Donovan, M. Springett,

T. Orszulak, D. A. Fullerton, A. J. Tajik, R. O. Bonow, and T. Spelsberg, "Human aortic

Page 83: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

73

valve calcification is associated with an osteoblast phenotype," Circulation, vol. 107, pp.

2181-4, May 6 2003.

[81] E. R. Mohler, 3rd, L. P. Adam, P. McClelland, L. Graham, and D. R. Hathaway,

"Detection of osteopontin in calcified human aortic valves," Arteriosclerosis Thrombosis

and Vascular Biology, vol. 17, pp. 547-552, Mar 1997.

[82] S. S. Srivatsa, P. J. Harrity, P. B. Maercklein, L. Kleppe, J. Veinot, W. D. Edwards, C.

M. Johnson, and L. A. Fitzpatrick, "Increased cellular expression of matrix proteins that

regulate mineralization is associated with calcification of native human and porcine

xenograft bioprosthetic heart valves," Journal of Clinical Investigation, vol. 99, pp. 996-

1009, Mar 1997.

[83] K. L. Weind, C. G. Ellis, and D. R. Boughner, "Aortic valve cusp vessel density:

Relationship with tissue thickness," Journal of Thoracic and Cardiovascular Surgery,

vol. 123, pp. 333-340, Feb 2002.

[84] D. Hakuno, N. Kimura, M. Yoshioka, M. Mukai, T. Kimura, Y. Okada, R. Yozu, C.

Shukunami, Y. Hiraki, A. Kudo, S. Ogawa, and K. Fukuda, "Periostin advances

atherosclerotic and rheumatic cardiac valve degeneration by inducing angiogenesis and

MMP production in humans and rodents," J Clin Invest, vol. 120, pp. 2292-306, Jul 2010.

[85] E. Nagy, P. Eriksson, M. Yousry, K. Caidahl, E. Ingelsson, G. K. Hansson, A. Franco-

Cereceda, and M. Back, "Valvular osteoclasts in calcification and aortic valve stenosis

severity," International Journal of Cardiology, vol. 168, pp. 2264-71, 2013 Oct 3 (Epub

2013 Feb 2013.

[86] I. Steiner, P. Kasparova, A. Kohout, and J. Dominik, "Bone formation in cardiac valves:

a histopathological study of 128 cases," Virchows Arch, vol. 450, pp. 653-7, Jun 2007.

[87] D. Magne, M. Julien, C. Vinatier, F. Merhi-Soussi, P. Weiss, and J. Guicheux, "Cartilage

formation in growth plate and arteries: from physiology to pathology," Bioessays, vol. 27,

pp. 708-16, Jul 2005.

[88] E. E. Wirrig and K. E. Yutzey, "Transcriptional regulation of heart valve development

and disease," Cardiovasc Pathol, vol. 20, pp. 162-7, May-Jun 2011.

[89] A. Alexopoulos, V. Bravou, S. Peroukides, L. Kaklamanis, J. Varakis, D. Alexopoulos,

and H. Papadaki, "Bone regulatory factors NFATc1 and Osterix in human calcific aortic

valves," Int J Cardiol, vol. 139, pp. 142-9, Mar 4 2010.

[90] H. Akiyama, M. C. Chaboissier, J. F. Martin, A. Schedl, and B. de Crombrugghe, "The

transcription factor Sox9 has essential roles in successive steps of the chondrocyte

differentiation pathway and is required for expression of Sox5 and Sox6," Genes Dev,

vol. 16, pp. 2813-28, Nov 1 2002.

[91] D. M. Bell, K. K. Leung, S. C. Wheatley, L. J. Ng, S. Zhou, K. W. Ling, M. H. Sham, P.

Koopman, P. P. Tam, and K. S. Cheah, "SOX9 directly regulates the type-II collagen

gene," Nat Genet, vol. 16, pp. 174-8, Jun 1997.

Page 84: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

74

[92] W. M. Bi, J. M. Deng, Z. P. Zhang, R. R. Behringer, and B. de Crombrugghe, "Sox9 is

required for cartilage formation," Nature Genetics, vol. 22, pp. 85-89, May 1999.

[93] J. J. Kaden, S. Bickelhaupt, R. Grobholz, C. F. Vahl, S. Hagl, M. Brueckmann, K. K.

Haase, C. E. Dempfle, and M. Borggrefe, "Expression of bone sialoprotein and bone

morphogenetic protein-2 in calcific aortic stenosis," J Heart Valve Dis, vol. 13, pp. 560-

6, Jul 2004.

[94] B. K. Zehentner, C. Dony, and H. Burtscher, "The transcription factor Sox9 is involved in

BMP-2 signaling," J Bone Miner Res, vol. 14, pp. 1734-41, Oct 1999.

[95] S. Murakami, M. Kan, W. L. McKeehan, and B. de Crombrugghe, "Up-regulation of the

chondrogenic Sox9 gene by fibroblast growth factors is mediated by the mitogen-

activated protein kinase pathway," Proc Natl Acad Sci U S A, vol. 97, pp. 1113-8, Feb 1

2000.

[96] M. A. Arnold, Y. Kim, M. P. Czubryt, D. Phan, J. McAnally, X. Qi, J. M. Shelton, J. A.

Richardson, R. Bassel-Duby, and E. N. Olson, "MEF2C transcription factor controls

chondrocyte hypertrophy and bone development," Dev Cell, vol. 12, pp. 377-89, Mar

2007.

[97] P. Bialek, B. Kern, X. Yang, M. Schrock, D. Sosic, N. Hong, H. Wu, K. Yu, D. M.

Ornitz, E. N. Olson, M. J. Justice, and G. Karsenty, "A twist code determines the onset of

osteoblast differentiation," Dev Cell, vol. 6, pp. 423-35, Mar 2004.

[98] I. Satokata, L. Ma, H. Ohshima, M. Bei, I. Woo, K. Nishizawa, T. Maeda, Y. Takano, M.

Uchiyama, S. Heaney, H. Peters, Z. Tang, R. Maxson, and R. Maas, "Msx2 deficiency in

mice causes pleiotropic defects in bone growth and ectodermal organ formation," Nat

Genet, vol. 24, pp. 391-5, Apr 2000.

[99] A. O. Wilkie, Z. Tang, N. Elanko, S. Walsh, S. R. Twigg, J. A. Hurst, S. A. Wall, K. H.

Chrzanowska, and R. E. Maxson, Jr., "Functional haploinsufficiency of the human

homeobox gene MSX2 causes defects in skull ossification," Nat Genet, vol. 24, pp. 387-

90, Apr 2000.

[100] T. Komori, H. Yagi, S. Nomura, A. Yamaguchi, K. Sasaki, K. Deguchi, Y. Shimizu, R.

T. Bronson, Y. H. Gao, M. Inada, M. Sato, R. Okamoto, Y. Kitamura, S. Yoshiki, and T.

Kishimoto, "Targeted disruption of Cbfa1 results in a complete lack of bone formation

owing to maturational arrest of osteoblasts," Cell, vol. 89, pp. 755-64, May 30 1997.

[101] Q. Zheng, G. Zhou, R. Morello, Y. Chen, X. Garcia-Rojas, and B. Lee, "Type X collagen

gene regulation by Runx2 contributes directly to its hypertrophic chondrocyte-specific

expression in vivo," J Cell Biol, vol. 162, pp. 833-42, Sep 1 2003.

[102] E. Serfling, S. Chuvpilo, J. Liu, T. Hofer, and A. Palmetshofer, "NFATc1 autoregulation:

a crucial step for cell-fate determination," Trends Immunol, vol. 27, pp. 461-9, Oct 2006.

Page 85: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

75

[103] T. Koga, Y. Matsui, M. Asagiri, T. Kodama, B. de Crombrugghe, K. Nakashima, and H.

Takayanagi, "NFAT and Osterix cooperatively regulate bone formation," Nat Med, vol.

11, pp. 880-5, Aug 2005.

[104] K. Nakashima, X. Zhou, G. Kunkel, Z. Zhang, J. M. Deng, R. R. Behringer, and B. de

Crombrugghe, "The novel zinc finger-containing transcription factor osterix is required

for osteoblast differentiation and bone formation," Cell, vol. 108, pp. 17-29, Jan 11 2002.

[105] J. L. Allen, M. E. Cooke, and T. Alliston, "ECM stiffness primes the TGFbeta pathway to

promote chondrocyte differentiation," Mol Biol Cell, vol. 23, pp. 3731-42, Sep 2012.

[106] H. J. Kwon and K. Yasuda, "Chondrogenesis on sulfonate-coated hydrogels is regulated

by their mechanical properties," J Mech Behav Biomed Mater, vol. 17, pp. 337-46, Jan

2013.

[107] T. Ikeda, S. Kamekura, A. Mabuchi, I. Kou, S. Seki, T. Takato, K. Nakamura, H.

Kawaguchi, S. Ikegawa, and U. I. Chung, "The combination of SOX5, SOX6, and SOX9

(the SOX trio) provides signals sufficient for induction of permanent cartilage," Arthritis

Rheum, vol. 50, pp. 3561-73, Nov 2004.

[108] Y. Nakashima, T. N. Wight, and K. Sueishi, "Early atherosclerosis in humans: role of

diffuse intimal thickening and extracellular matrix proteoglycans," Cardiovasc Res, vol.

79, pp. 14-23, Jul 1 2008.

[109] Y. Nakashima, Y. X. Chen, N. Kinukawa, and K. Sueishi, "Distributions of diffuse

intimal thickening in human arteries: preferential expression in atherosclerosis-prone

arteries from an early age," Virchows Arch, vol. 441, pp. 279-88, Sep 2002.

[110] M. J. Merrilees, B. Beaumont, and L. J. Scott, "Comparison of deposits of versican,

biglycan and decorin in saphenous vein and internal thoracic, radial and coronary

arteries: correlation to patency," Coron Artery Dis, vol. 12, pp. 7-16, Feb 2001.

[111] L. R. Tannock and V. L. King, "Proteoglycan mediated lipoprotein retention: a

mechanism of diabetic atherosclerosis," Rev Endocr Metab Disord, vol. 9, pp. 289-300,

Dec 2008.

[112] I. Tabas, K. J. Williams, and J. Boren, "Subendothelial lipoprotein retention as the

initiating process in atherosclerosis: update and therapeutic implications," Circulation,

vol. 116, pp. 1832-44, Oct 16 2007.

[113] K. L. Olin, S. Potter-Perigo, P. H. Barrett, T. N. Wight, and A. Chait, "Lipoprotein lipase

enhances the binding of native and oxidized low density lipoproteins to versican and

biglycan synthesized by cultured arterial smooth muscle cells," J Biol Chem, vol. 274, pp.

34629-36, Dec 3 1999.

[114] M. O. Pentikainen, K. Oorni, and P. T. Kovanen, "Lipoprotein lipase (LPL) strongly links

native and oxidized low density lipoprotein particles to decorin-coated collagen. Roles

for both dimeric and monomeric forms of LPL," J Biol Chem, vol. 275, pp. 5694-701,

Feb 25 2000.

Page 86: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

76

[115] M. Gustafsson, M. Levin, K. Skalen, J. Perman, V. Friden, P. Jirholt, S. O. Olofsson, S.

Fazio, M. F. Linton, C. F. Semenkovich, G. Olivecrona, and J. Boren, "Retention of low-

density lipoprotein in atherosclerotic lesions of the mouse: evidence for a role of

lipoprotein lipase," Circ Res, vol. 101, pp. 777-83, Oct 12 2007.

[116] J. E. Figueroa and P. Vijayagopal, "Angiotensin II stimulates synthesis of vascular

smooth muscle cell proteoglycans with enhanced low density lipoprotein binding

properties," Atherosclerosis, vol. 162, pp. 261-8, Jun 2002.

[117] P. J. Little, L. Tannock, K. L. Olin, A. Chait, and T. N. Wight, "Proteoglycans

synthesized by arterial smooth muscle cells in the presence of transforming growth

factor-beta1 exhibit increased binding to LDLs," Arterioscler Thromb Vasc Biol, vol. 22,

pp. 55-60, Jan 2002.

[118] C. D. Meyers, L. R. Tannock, T. N. Wight, and A. Chait, "Statin-exposed vascular

smooth muscle cells secrete proteoglycans with decreased binding affinity for LDL," J

Lipid Res, vol. 44, pp. 2152-60, Nov 2003.

[119] K. Hashimura, K. Sudhir, J. Nigro, S. Ling, M. R. Williams, P. A. Komesaroff, and P. J.

Little, "Androgens stimulate human vascular smooth muscle cell proteoglycan

biosynthesis and increase lipoprotein binding," Endocrinology, vol. 146, pp. 2085-90,

Apr 2005.

[120] K. D. O'Brien, K. Lewis, J. W. Fischer, P. Johnson, J. Y. Hwang, E. A. Knopp, M. G.

Kinsella, P. H. Barrett, A. Chait, and T. N. Wight, "Smooth muscle cell biglycan

overexpression results in increased lipoprotein retention on extracellular matrix:

implications for the retention of lipoproteins in atherosclerosis," Atherosclerosis, vol.

177, pp. 29-35, Nov 2004.

[121] K. Skalen, M. Gustafsson, E. K. Rydberg, L. M. Hulten, O. Wiklund, T. L. Innerarity,

and J. Boren, "Subendothelial retention of atherogenic lipoproteins in early

atherosclerosis," Nature, vol. 417, pp. 750-4, Jun 13 2002.

[122] A. H. Rapp, L. D. Hillis, R. A. Lange, and J. E. Cigarroa, "Prevalence of coronary artery

disease in patients with aortic stenosis with and without angina pectoris," Am J Cardiol,

vol. 87, pp. 1216-7; A7, May 15 2001.

[123] S. R. Srinivasan, P. Vijayagopal, K. Eberle, B. Radhakrishnamurthy, and G. S. Berenson,

"Low-density lipoprotein binding affinity of arterial wall proteoglycans: characteristics of

a chondroitin sulfate proteoglycan subfraction," Biochim Biophys Acta, vol. 1006, pp.

159-66, Nov 28 1989.

[124] S. R. Srinivasan, P. Vijayagopal, K. Eberle, E. R. Dalferes, Jr., B. Radhakrishnamurthy,

and G. S. Berenson, "Low density lipoprotein binding affinity of arterial wall isomeric

chondroitin sulfate proteoglycans," Atherosclerosis, vol. 72, pp. 1-9, Jul 1988.

[125] M. T. Dours-Zimmermann and D. R. Zimmermann, "A novel glycosaminoglycan

attachment domain identified in two alternative splice variants of human versican," J Biol

Chem, vol. 269, pp. 32992-8, Dec 30 1994.

Page 87: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

77

[126] N. Osman, K. J. Grande-Allen, M. L. Ballinger, R. Getachew, S. Marasco, K. D. O'Brien,

and P. J. Little, "Smad2-dependent glycosaminoglycan elongation in aortic valve

interstitial cells enhances binding of LDL to proteoglycans," Cardiovasc Pathol, vol. 22,

pp. 146-55, Mar-Apr 2013.

[127] T. N. Wight and M. J. Merrilees, "Proteoglycans in atherosclerosis and restenosis: key

roles for versican," Circ Res, vol. 94, pp. 1158-67, May 14 2004.

[128] K. Olin-Lewis, J. L. Benton, J. C. Rutledge, D. G. Baskin, T. N. Wight, and A. Chait,

"Apolipoprotein E mediates the retention of high-density lipoproteins by mouse carotid

arteries and cultured arterial smooth muscle cell extracellular matrices," Circ Res, vol.

90, pp. 1333-9, Jun 28 2002.

[129] H. Derbali, Y. Bosse, N. Cote, P. Pibarot, A. Audet, A. Pepin, B. Arsenault, C. Couture,

J. P. Despres, and P. Mathieu, "Increased biglycan in aortic valve stenosis leads to the

overexpression of phospholipid transfer protein via Toll-like receptor 2," Am J Pathol,

vol. 176, pp. 2638-45, Jun 2010.

[130] R. Song, Q. Zeng, L. Ao, J. A. Yu, J. C. Cleveland, K. S. Zhao, D. A. Fullerton, and X.

Meng, "Biglycan induces the expression of osteogenic factors in human aortic valve

interstitial cells via Toll-like receptor-2," Arterioscler Thromb Vasc Biol, vol. 32, pp.

2711-20, Nov 2012.

[131] C. C. Reed and R. V. Iozzo, "The role of decorin in collagen fibrillogenesis and skin

homeostasis," Glycoconj J, vol. 19, pp. 249-55, May-Jun 2002.

[132] G. A. Walker, K. S. Masters, D. N. Shah, K. S. Anseth, and L. A. Leinwand, "Valvular

myofibroblast activation by transforming growth factor-beta: implications for

pathological extracellular matrix remodeling in heart valve disease," Circ Res, vol. 95,

pp. 253-60, Aug 6 2004.

[133] M. Pho, W. Lee, D. R. Watt, C. Laschinger, C. A. Simmons, and C. A. McCulloch,

"Cofilin is a marker of myofibroblast differentiation in cells from porcine aortic cardiac

valves," Am J Physiol Heart Circ Physiol, vol. 294, pp. H1767-78, Apr 2008.

[134] J. N. Clark-Greuel, J. M. Connolly, E. Sorichillo, N. R. Narula, H. S. Rapoport, E. R.

Mohler, 3rd, J. H. Gorman, 3rd, R. C. Gorman, and R. J. Levy, "Transforming growth

factor-beta1 mechanisms in aortic valve calcification: increased alkaline phosphatase and

related events," Ann Thorac Surg, vol. 83, pp. 946-53, Mar 2007.

[135] B. Johansson, A. Holmgren, M. Hedstrom, A. Engstrom-Laurent, and K. G. Engstrom,

"Evaluation of hyaluronan and calcifications in stenotic and regurgitant aortic valves,"

Eur J Cardiothorac Surg, vol. 39, pp. 27-32, Jan 2011.

[136] T. Ito, J. D. Williams, D. Fraser, and A. O. Phillips, "Hyaluronan attenuates transforming

growth factor-beta1-mediated signaling in renal proximal tubular epithelial cells," Am J

Pathol, vol. 164, pp. 1979-88, Jun 2004.

Page 88: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

78

[137] C. A. Cuff, D. Kothapalli, I. Azonobi, S. Chun, Y. Zhang, R. Belkin, C. Yeh, A. Secreto,

R. K. Assoian, D. J. Rader, and E. Pure, "The adhesion receptor CD44 promotes

atherosclerosis by mediating inflammatory cell recruitment and vascular cell activation,"

J Clin Invest, vol. 108, pp. 1031-40, Oct 2001.

[138] S. P. Evanko, E. W. Raines, R. Ross, L. I. Gold, and T. N. Wight, "Proteoglycan

distribution in lesions of atherosclerosis depends on lesion severity, structural

characteristics, and the proximity of platelet-derived growth factor and transforming

growth factor-beta," Am J Pathol, vol. 152, pp. 533-46, Feb 1998.

[139] S. P. Evanko, J. C. Angello, and T. N. Wight, "Formation of hyaluronan- and versican-

rich pericellular matrix is required for proliferation and migration of vascular smooth

muscle cells," Arterioscler Thromb Vasc Biol, vol. 19, pp. 1004-13, Apr 1999.

[140] E. H. Stephens, J. G. Saltarrelli, Jr., L. R. Balaoing, L. S. Baggett, I. Nandi, K. M.

Anderson, J. D. Morrisett, M. J. Reardon, M. A. Simpson, P. H. Weigel, E. A. Olmsted-

Davis, A. R. Davis, and K. J. Grande-Allen, "Hyaluronan turnover and hypoxic brown

adipocytic differentiation are co-localized with ossification in calcified human aortic

valves," Pathol Res Pract, vol. 208, pp. 642-50, Nov 15 2012.

[141] G. Embery, S. Rees, R. Hall, K. Rose, R. Waddington, and P. Shellis, "Calcium- and

hydroxyapatite-binding properties of glucuronic acid-rich and iduronic acid-rich

glycosaminoglycans and proteoglycans," Eur J Oral Sci, vol. 106 Suppl 1, pp. 267-73,

Jan 1998.

[142] K. L. Sider, M. C. Blaser, and C. A. Simmons, "Animal models of calcific aortic valve

disease," Int J Inflam, vol. 2011, p. 364310, 2011.

[143] J. R. Turk and M. H. Laughlin, "Physical activity and atherosclerosis: which animal

model?," Can J Appl Physiol, vol. 29, pp. 657-83, Oct 2004.

[144] M. H. Moghadasian, J. J. Frohlich, and B. M. McManus, "Advances in experimental

dyslipidemia and atherosclerosis," Lab Invest, vol. 81, pp. 1173-83, Sep 2001.

[145] M. Guerraty and E. R. Mohler, 3rd, "Models of aortic valve calcification," J Investig

Med, vol. 55, pp. 278-83, Sep 2007.

[146] R. G. Gerrity, R. Natarajan, J. L. Nadler, and T. Kimsey, "Diabetes-induced accelerated

atherosclerosis in swine," Diabetes, vol. 50, pp. 1654-65, Jul 2001.

[147] R. M. Weiss, M. Ohashi, J. D. Miller, S. G. Young, and D. D. Heistad, "Calcific aortic

valve stenosis in old hypercholesterolemic mice," Circulation, vol. 114, pp. 2065-9, Nov

7 2006.

[148] K. Tanaka, M. Sata, D. Fukuda, Y. Suematsu, N. Motomura, S. Takamoto, Y. Hirata, and

R. Nagai, "Age-associated aortic stenosis in apolipoprotein E-deficient mice," J Am Coll

Cardiol, vol. 46, pp. 134-41, Jul 5 2005.

Page 89: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

79

[149] M. C. Drolet, E. Roussel, Y. Deshaies, J. Couet, and M. Arsenault, "A high fat/high

carbohydrate diet induces aortic valve disease in C57BL/6J mice," J Am Coll Cardiol,

vol. 47, pp. 850-5, Feb 21 2006.

[150] M. Nus, D. MacGrogan, B. Martinez-Poveda, Y. Benito, J. C. Casanova, F. Fernandez-

Aviles, J. Bermejo, and J. L. de la Pompa, "Diet-induced aortic valve disease in mice

haploinsufficient for the Notch pathway effector RBPJK/CSL," Arterioscler Thromb

Vasc Biol, vol. 31, pp. 1580-8, Jul 2011.

[151] M. C. Drolet, M. Arsenault, and J. Couet, "Experimental aortic valve stenosis in rabbits,"

J Am Coll Cardiol, vol. 41, pp. 1211-7, Apr 2 2003.

[152] M. C. Drolet, J. Couet, and M. Arsenault, "Development of aortic valve sclerosis or

stenosis in rabbits: role of cholesterol and calcium," J Heart Valve Dis, vol. 17, pp. 381-

7, Jul 2008.

[153] N. M. Rajamannan, G. Sangiorgi, M. Springett, K. Arnold, T. Mohacsi, L. G. Spagnoli,

W. D. Edwards, A. J. Tajik, and R. S. Schwartz, "Experimental hypercholesterolemia

induces apoptosis in the aortic valve," J Heart Valve Dis, vol. 10, pp. 371-4, May 2001.

[154] D. W. Swinkels and P. N. Demacker, "Comparative studies on the low density

lipoprotein subfractions from pig and man," Comp Biochem Physiol B, vol. 90, pp. 297-

300, 1988.

[155] J. L. Dixon, J. D. Stoops, J. L. Parker, M. H. Laughlin, G. A. Weisman, and M. Sturek,

"Dyslipidemia and vascular dysfunction in diabetic pigs fed an atherogenic diet,"

Arterioscler Thromb Vasc Biol, vol. 19, pp. 2981-92, Dec 1999.

[156] Guide for the Care and Use of Laboratory Animals. Washington DC: National Academy

of Sciences, 2011.

[157] M. O. Pentikainen, K. Oorni, R. Lassila, and P. T. Kovanen, "The proteoglycan decorin

links low density lipoproteins with collagen type I," J Biol Chem, vol. 272, pp. 7633-8,

Mar 21 1997.

[158] C. Wiberg, A. R. Klatt, R. Wagener, M. Paulsson, J. F. Bateman, D. Heinegard, and M.

Morgelin, "Complexes of matrilin-1 and biglycan or decorin connect collagen VI

microfibrils to both collagen II and aggrecan," J Biol Chem, vol. 278, pp. 37698-704, Sep

26 2003.

[159] K. D. O'Brien, S. Vuletic, T. O. McDonald, G. Wolfbauer, K. Lewis, A. Y. Tu, S.

Marcovina, T. N. Wight, A. Chait, and J. J. Albers, "Cell-associated and extracellular

phospholipid transfer protein in human coronary atherosclerosis," Circulation, vol. 108,

pp. 270-4, Jul 22 2003.

[160] J. S. Park, J. S. Chu, A. D. Tsou, R. Diop, Z. Tang, A. Wang, and S. Li, "The effect of

matrix stiffness on the differentiation of mesenchymal stem cells in response to TGF-

beta," Biomaterials, vol. 32, pp. 3921-30, Jun 2011.

Page 90: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

80

[161] I. El-Hamamsy, K. Balachandran, M. H. Yacoub, L. M. Stevens, P. Sarathchandra, P. M.

Taylor, A. P. Yoganathan, and A. H. Chester, "Endothelium-dependent regulation of the

mechanical properties of aortic valve cusps," J Am Coll Cardiol, vol. 53, pp. 1448-55,

Apr 21 2009.

[162] J. Richards, I. El-Hamamsy, S. Chen, Z. Sarang, P. Sarathchandra, M. H. Yacoub, A. H.

Chester, and J. T. Butcher, "Side-specific endothelial-dependent regulation of aortic

valve calcification: interplay of hemodynamics and nitric oxide signaling," Am J Pathol,

vol. 182, pp. 1922-31, May 2013.

[163] S. T. Gould, E. E. Matherly, J. N. Smith, D. D. Heistad, and K. S. Anseth, "The role of

valvular endothelial cell paracrine signaling and matrix elasticity on valvular interstitial

cell activation," Biomaterials, vol. 35, pp. 3596-606, Apr 2014.

[164] J. H. Chen, C. Y. Yip, E. D. Sone, and C. A. Simmons, "Identification and

characterization of aortic valve mesenchymal progenitor cells with robust osteogenic

calcification potential," Am J Pathol, vol. 174, pp. 1109-19, Mar 2009.

[165] K. Balachandran, P. Sucosky, H. Jo, and A. P. Yoganathan, "Elevated cyclic stretch alters

matrix remodeling in aortic valve cusps: implications for degenerative aortic valve

disease," Am J Physiol Heart Circ Physiol, vol. 296, pp. H756-64, Mar 2009.

[166] K. Balachandran, P. Sucosky, H. Jo, and A. P. Yoganathan, "Elevated cyclic stretch

induces aortic valve calcification in a bone morphogenic protein-dependent manner," Am

J Pathol, vol. 177, pp. 49-57, Jul 2010.

[167] P. Thayer, K. Balachandran, S. Rathan, C. H. Yap, S. Arjunon, H. Jo, and A. P.

Yoganathan, "The effects of combined cyclic stretch and pressure on the aortic valve

interstitial cell phenotype," Ann Biomed Eng, vol. 39, pp. 1654-67, Jun 2011.

[168] I. Vesely and R. Noseworthy, "Micromechanics of the fibrosa and the ventricularis in

aortic valve leaflets," J Biomech, vol. 25, pp. 101-13, Jan 1992.

[169] J. A. Stella and M. S. Sacks, "On the biaxial mechanical properties of the layers of the

aortic valve leaflet," J Biomech Eng, vol. 129, pp. 757-66, Oct 2007.

[170] R. Zhao, K. L. Sider, and C. A. Simmons, "Measurement of layer-specific mechanical

properties in multilayered biomaterials by micropipette aspiration," Acta Biomater, vol.

7, pp. 1220-7, Mar 2011.

[171] Y. Bosse, A. Miqdad, D. Fournier, A. Pepin, P. Pibarot, and P. Mathieu, "Refining

molecular pathways leading to calcific aortic valve stenosis by studying gene expression

profile of normal and calcified stenotic human aortic valves," Circ Cardiovasc Genet,

vol. 2, pp. 489-98, Oct 2009.

[172] V. Espina, J. D. Wulfkuhle, V. S. Calvert, A. VanMeter, W. Zhou, G. Coukos, D. H.

Geho, E. F. Petricoin, 3rd, and L. A. Liotta, "Laser-capture microdissection," Nat Protoc,

vol. 1, pp. 586-603, 2006.

Page 91: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

81

[173] J. C. Oliveros, "VENNY. An interactive tool for comparing lists with Venn Diagrams.,"

ed, 2007.

[174] X. Meng, L. Ao, Y. Song, A. Babu, X. Yang, M. Wang, M. J. Weyant, C. A. Dinarello, J.

C. Cleveland, Jr., and D. A. Fullerton, "Expression of functional Toll-like receptors 2 and

4 in human aortic valve interstitial cells: potential roles in aortic valve inflammation and

stenosis," American Journal of Physiology - Cell Physiology, vol. 294, pp. C29-C35, Jan

2008.

[175] E. E. Wirrig and K. E. Yutzey, "Developmental Pathways in CAVD," in Calcific Aortic

Valve Disease, E. Aikawa, Ed., ed: InTech, 2013.

[176] A. Fischer, C. Steidl, T. U. Wagner, E. Lang, P. M. Jakob, P. Friedl, K. P. Knobeloch,

and M. Gessler, "Combined loss of Hey1 and HeyL causes congenital heart defects

because of impaired epithelial to mesenchymal transition," Circ Res, vol. 100, pp. 856-

63, Mar 30 2007.

[177] B. Chen, R. T. Bronson, L. D. Klaman, T. G. Hampton, J. F. Wang, P. J. Green, T.

Magnuson, P. S. Douglas, J. P. Morgan, and B. G. Neel, "Mice mutant for Egfr and Shp2

have defective cardiac semilunar valvulogenesis," Nat Genet, vol. 24, pp. 296-9, Mar

2000.

[178] K. L. McBride, G. A. Zender, S. M. Fitzgerald-Butt, N. J. Seagraves, S. D. Fernbach, G.

Zapata, M. Lewin, J. A. Towbin, and J. W. Belmont, "Association of common variants in

ERBB4 with congenital left ventricular outflow tract obstruction defects," Birth Defects

Res A Clin Mol Teratol, vol. 91, pp. 162-8, Mar 2011.

[179] C. Centers for Disease and Prevention, "Hospital stays, hospital charges, and in-hospital

deaths among infants with selected birth defects--United States, 2003," MMWR Morb

Mortal Wkly Rep, vol. 56, pp. 25-9, Jan 19 2007.

[180] W. K. Clements, A. D. Kim, K. G. Ong, J. C. Moore, N. D. Lawson, and D. Traver, "A

somitic Wnt16/Notch pathway specifies haematopoietic stem cells," Nature, vol. 474, pp.

220-4, Jun 9 2011.

[181] R. L. Wilensky, Y. Shi, E. R. Mohler, 3rd, D. Hamamdzic, M. E. Burgert, J. Li, A.

Postle, R. S. Fenning, J. G. Bollinger, B. E. Hoffman, D. J. Pelchovitz, J. Yang, R. C.

Mirabile, C. L. Webb, L. Zhang, P. Zhang, M. H. Gelb, M. C. Walker, A. Zalewski, and

C. H. Macphee, "Inhibition of lipoprotein-associated phospholipase A2 reduces complex

coronary atherosclerotic plaque development," Nat Med, vol. 14, pp. 1059-66, Oct 2008.

[182] A. Mahmut, M. C. Boulanger, D. El Husseini, D. Fournier, R. Bouchareb, J. P. Despres,

P. Pibarot, Y. Bosse, and P. Mathieu, "Elevated expression of lipoprotein-associated

phospholipase A2 in calcific aortic valve disease: implications for valve mineralization,"

J Am Coll Cardiol, vol. 63, pp. 460-9, Feb 11 2014.

[183] D. El Husseini, M. C. Boulanger, D. Fournier, A. Mahmut, Y. Bosse, P. Pibarot, and P.

Mathieu, "High expression of the Pi-transporter SLC20A1/Pit1 in calcific aortic valve

Page 92: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

82

disease promotes mineralization through regulation of Akt-1," PLoS One, vol. 8, p.

e53393, 2013.

[184] J. D. Peacock, A. K. Levay, D. B. Gillaspie, G. Tao, and J. Lincoln, "Reduced sox9

function promotes heart valve calcification phenotypes in vivo," Circ Res, vol. 106, pp.

712-9, Mar 5 2010.

[185] X. Yang, D. A. Fullerton, X. Su, L. Ao, J. C. Cleveland, Jr., and X. Meng, "Pro-

osteogenic phenotype of human aortic valve interstitial cells is associated with higher

levels of Toll-like receptors 2 and 4 and enhanced expression of bone morphogenetic

protein 2," J Am Coll Cardiol, vol. 53, pp. 491-500, Feb 10 2009.

[186] Y. Song, D. A. Fullerton, D. Mauchley, X. Su, L. Ao, X. Yang, J. C. Cleveland, and X.

Meng, "Microfilaments facilitate TLR4-mediated ICAM-1 expression in human aortic

valve interstitial cells," J Surg Res, vol. 166, pp. 52-8, Mar 2011.

[187] D. J. Samuvel, K. P. Sundararaj, Y. Li, M. F. Lopes-Virella, and Y. Huang, "Adipocyte-

mononuclear cell interaction, Toll-like receptor 4 activation, and high glucose

synergistically up-regulate osteopontin expression via an interleukin 6-mediated

mechanism," J Biol Chem, vol. 285, pp. 3916-27, Feb 5 2010.

[188] G. P. Shi, G. K. Sukhova, M. Kuzuya, Q. Ye, J. Du, Y. Zhang, J. H. Pan, M. L. Lu, X. W.

Cheng, A. Iguchi, S. Perrey, A. M. Lee, H. A. Chapman, and P. Libby, "Deficiency of the

cysteine protease cathepsin S impairs microvessel growth," Circ Res, vol. 92, pp. 493-

500, Mar 21 2003.

[189] S. Helske, S. Syvaranta, K. A. Lindstedt, J. Lappalainen, K. Oorni, M. I. Mayranpaa, J.

Lommi, H. Turto, K. Werkkala, M. Kupari, and P. T. Kovanen, "Increased expression of

elastolytic cathepsins S, K, and V and their inhibitor cystatin C in stenotic aortic valves,"

Arterioscler Thromb Vasc Biol, vol. 26, pp. 1791-8, Aug 2006.

[190] Q. Zeng, R. Song, L. Ao, D. Xu, N. Venardos, D. A. Fullerton, and X. Meng,

"Augmented Osteogenic Responses in Human Aortic Valve Cells Exposed to oxLDL and

TLR4 Agonist: A Mechanistic Role of Notch1 and NF-kappaB Interaction," PLoS One,

vol. 9, p. e95400, 2014.

[191] E. A. Turley, P. W. Noble, and L. Y. Bourguignon, "Signaling properties of hyaluronan

receptors," J Biol Chem, vol. 277, pp. 4589-92, Feb 15 2002.

[192] S. V. Kyosseva, E. N. Harris, and P. H. Weigel, "The hyaluronan receptor for endocytosis

mediates hyaluronan-dependent signal transduction via extracellular signal-regulated

kinases," J Biol Chem, vol. 283, pp. 15047-55, May 30 2008.

[193] C. M. McKee, M. B. Penno, M. Cowman, M. D. Burdick, R. M. Strieter, C. Bao, and P.

W. Noble, "Hyaluronan (HA) fragments induce chemokine gene expression in alveolar

macrophages. The role of HA size and CD44," J Clin Invest, vol. 98, pp. 2403-13, Nov

15 1996.

Page 93: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

83

[194] P. W. Noble, C. M. McKee, M. Cowman, and H. S. Shin, "Hyaluronan fragments activate

an NF-kappa B/I-kappa B alpha autoregulatory loop in murine macrophages," J Exp Med,

vol. 183, pp. 2373-8, May 1 1996.

[195] D. C. West, I. N. Hampson, F. Arnold, and S. Kumar, "Angiogenesis induced by

degradation products of hyaluronic acid," Science, vol. 228, pp. 1324-6, Jun 14 1985.

Page 94: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

84

Appendices

A. Supplemental Data

A.1 Myofibroblast detection in porcine valve lesions

Previously, myofibroblast absence in early PG-rich lesions was detected using

immunoperoxidase methods by Sider et al. (2013). Specifically, no myofibroblasts were

observed at two months, while only few were observed at the base of lesions from the five-

month time point. Immunofluorescence (IF) provides greater signal sensitivity and was used to

corroborate the results from immunoperoxidase staining.

Materials and Methods

Formalin-fixed, paraffin-embedded radial centre sections from the early porcine model by Sider

et al. (2013) were previously stained for alph-smooth muscle actin (αSMA) using

immunoperoxidase methods. For direct comparison, serial sections were used for IF staining.

Briefly, IF staining began by melting the wax on sections at 60°C for 30 min. This was followed

by deparaffinization in xylene and rehydration in graded ethanol baths. Antigen retrieval

involved incubation in 1 µg/µl Trypsin-CaCl2 (Sigma-Aldrich) for 30 min in a 37°C water bath.

Non-specific staining was blocking using 10% goat serum buffer for 45 min prior to an hour-

long incubation with anti-αSMA primary antibody (mouse monoclonal, 4 µg/mL, ab7817,

Abcam). Samples were then incubated for 30 min with Alexa Fluor 568 goat anti-mouse

antibody (A-11004, 20 µg/mL, Life Technologies, Burlington, ON) diluted in 10% goat serum

buffer. Nuclei were stained with Hoechst (33258, 1µg/mL, Sigma-Aldrich) for 5 min.

PBS/Tween was used to wash between steps. Negative controls involved no primary and mouse

IgG controls (Santa Cruz).

Results and Discussion

PG-rich lesions of interest were visualized using Movat’s pentachrome. IF staining did not

identify any additional lesions with αSMA-positive cells that were not previously detected by

immunoperoxidase staining (Figure A.1). In the few lesions that did demonstrate areas of

positive αSMA, staining was spatially similar between immunoperoxidase and IF stained serial

sections.

Page 95: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

85

(A)

(B)

(C)

(D)

(E)

(F)

Figure A.1. Immunoperoxidase (IP) and immunofluorescent (IF) staining of alpha-smooth muscle actin

(αSMA). IF did not identify αSMA positive lesions in addition to those found by IP staining (A,C,E). Staining

patterns of positive areas were similar in IP and IF staining (B,D,F). MP = Movat’s pentachrome (blue =

proteoglycan, yellow = collagen, black = nuclei/elastin, red = cytoplasm). Scale bar = 60µm.

MP MP

αSMA IP αSMA IP

αSMA IF αSMA IF

Page 96: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

86

A.2 Porcine model diet formulation

The porcine model was designed and prepared by Sider et al. (2013). Barrows were supplied by

the Arkell Swine Research Station, University of Guelph. The porcine diet was formulated based

on the standard Guelph University Arkell Swine Research diets and simplified to a corn and

soybean base. All diets were balanced for protein, carbohydrate, fat, digestible lysine, calcium,

and available phosphates. Diets are isocaloric between control and experimental diets when

experimental diets are fed at 87% of control diet levels. Protein kcal% between control and

experimental diets were equalized, as were carbohydrate + fat kcal% between diets. The starter

diets were fed up until swine reached ~40kg, at which point they were fed the grower diets until

the study ended. All diets were verified by Dr. C. Kees De Lange [Animal and Poultry Science,

University of Guelph]. All diet components were supplied by Arkell other than the cholesterol

[Research Diets, Inc., New Brunswick, NJ, USA], and liquid lard [Quality Meat Packers, Ltd.,

Toronto, ON, Canada]. Diets were milled at the Arkell Research Feed Mill, University of

Guelph.

Table A.1. Control starter III diet formulation

Page 97: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

87

Table A.2. Experimental starter III (12% lard, 1.5% cholesterol) diet formulation

Table A.3. Control grower diet formulation

Table A.4. Experimental grower (12% lard, 1.5% cholesterol) diet formulation

Page 98: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

88

A.3 PG/GAG scoring validation and analyses

Lesions stained for biglycan, decorin, versican, and hyaluronan were divided into regions of

interest (ROIs) to allow for localized PG/GAG correlations. Lesions images were divided into

grids with 100µm x 100µm ROIs to allow for more localized analysis of lesion areas. Each ROI

was semi-quanitatively scored for each PG/GAG and ApoB (Figure 3.2). To validate the results,

a lesion score was calculated and correlated to its corresponding percent PG/GAG staining of

total lesion (Figure A.2). Each ROI score was normalized to the fraction of lesion area in each

cell. The lesion score was calculated by adding all normalized ROI scores in a lesion and

dividing it by the total lesion area.

Results

According to Spearman’s correlation, the lesion scores for each PG/GAG strongly correlated

with the percent PG/GAG staining of total lesion (p<0.001).

(A)

(B)

(C)

(D)

Figure A.2. Semi-quantiative scores strongly correlate with PG/GAG staining of total lesion area

percentages.

Page 99: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

89

Using Kruskal Wallis tests with Mann-Whitney post-hoc tests with Bonferroni corrections

(p<0.0083), comparisons of PG/GAG lesion scores between pig time points and diet groups

yielded similar results to comparisons of percent area staining of PG/GAGs (Chapter 4). At two

months, lesions from the HF/HC diet have increased biglycan scores compared to lesions from

the normal chow diet (p<0.001). Furthermore, in HF/HC pigs, lesions from 5-months have lower

lesion scores of biglycan (p<0.001) and decorin (p=0.001) than those from 2-months.

Figure A.3. HF/HC diet alters lesion PG score with temporal differences. HF/HC = high-fat/high-

cholesterol diet. Error bars = SEM. *p<0.0083

A.4 Specific PG/GAG-rich lesions display distinct morphological

characteristics

Lesions rich in each PG/GAG were analyzed for association with ApoB-positive areas and Sox9-

expressing cells using Pearson’s Chi-Square Test or Fisher’s Exact Test. The latter was used in

the place of the former if one of the groups had a count of less than five. “Rich” lesions were

defined as those that express at least 45% PG/GAG staining of the total lesion area, as this is

approximately one-half standard deviations (±12.5%) above the mean percent PG/GAG staining

of the total lesion area for all PGs/GAG together (31.9%). ApoB positive lesions were defined as

those containing areas with a minimum score of one. Sox9-positive lesions were identified as

those with over ten percent Sox9 fraction. Spearman’s rank order correlation was also used to

correlate levels of percent staining of total lesion area between each of the PGs/GAG.

* *

*

Page 100: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

90

Results and Discussion

Lesions categorized by those rich with and those not rich with specific PGs/GAG were

characterized by several factors, including dense/diffuse morphology, ApoB presence/absence,

presence/absence of Sox9-expressing cells, using Pearson’s Chi-Square Test and Fisher’s Exact

Test (Table A.4). Biglycan-rich lesions (n=11) demonstrated distinct characteristics compared to

lesions that do not express high levels of biglycan. Firstly, they were primarily found in lesions

from swine fed the HF/HC diet (p=0.016). Furthermore, the majority of biglycan-rich lesions

were morphologically dense (p=0.001). Association with ApoB-positive regions (p=0.017) and

Sox9-expressing cells (p=0.009) was another common trait of these lesions. Similar to biglycan,

lesions rich in decorin (n=14) and versican (n=25) were more commonly observed in two month

old swine (p=0.008 and p=0.003, respectively). Decorin-rich lesions were also more commonly

associated with ApoB-positive regions (p=0.029), suggesting they may also play a role in lipid

retention. Interestingly, versican-rich lesions were also often biglycan-rich, and vice versa

(p=0.037). Hyaluronan-rich lesions (n=16) were less commonly observed in swine fed the

HF/HC diet (p=0.009). Further, these lesions were also less commonly associated with ApoB-

positive regions (p=0.035).

Table A.4. Qualitative categorization of specific PG/GAG-rich lesions

HF/HC

Diet

Two-Month

Time Point

Dense

Morphology

ApoB

+

Sox9

+

Richness in other

PG/GAGs

Biglycan-rich 10/11* 11/11* 10/11* 8/11* 11/11* 6/11 decorin-rich

9/11 versican-rich*

2/11 hyaluronan-rich

Decorin-rich 9/14 13/14* 8/14 9/14* 9/14 6/14 biglycan-rich

9/14 versican-rich

5/14 hyaluronan-rich

Versican-rich 16/25 21/25* 14/25 10/25 18/25 9/25 biglycan-rich*

9/25 decorin-rich

9/25 hyaluronan-rich

Hyaluronan-rich 5/16* 12/16* 4/16 3/16* 11/16 2/16 biglycan-rich

5/16 decorin-rich

9/16 versican-rich

*p<0.05, compared to all lesions that are not rich in the specified PG/GAG

A.5 RNA and cDNA quality control before microarray analysis

Valve interstitial cells (VICs) were isolated from dense lesions from four 2-month high-fat/high-

cholesterol (HF/HC) pigs. Using laser capture microdissection (LCM), three areas of interest

Page 101: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

91

were isolated from each sample: (1) top of the lesion; (2) bottom of the lesion; and (3) normal

fibrosa. RNA and cDNA quality were analyzed using a 2100 Bioanalyzer (Agilent Technologies

Canada Inc., Mississauga, ON, Canada) following RNA isolation and amplification, respectively.

RNA quality was examined using RNA integrity numbers (RINs) and noting the presence of

distinct peaks for 18S and 28S ribosomal subunits. Partial digestion was present in these RNA

samples. cDNA profiles indicated curves that plateaued around 200-500 nt with some high

molecular weight products around 500-1000 nt.

(A)

(B)

(C)

(D)

(E)

(F)

(G)

(H)

Figure A.4. Bioanalyzer results for top of lesion samples. RNA (A,C,E,G) and cDNA (B,D,F,H)

profiles indicate suitable samples for downstream microarray analysis. Pig Identifiers: (A) and (B) =

11107; (C) and (D) = Pig 11212; (E) and (F) = 12509; (G) and (H) = 11907. RIN = RNA Integrity

Number.

RIN = 5.4

RIN = 5.1

RIN = 6.2

RIN = 7.0

Page 102: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

92

(A)

(B)

(C)

(D)

(E)

(F)

(G)

(H)

Figure A.4. Bioanalyzer results for bottom of lesion samples. RNA (A,C,E,G) and cDNA (B,D,F,H)

profiles indicate suitable samples for downstream microarray analysis. Pig Identifiers: (A) and (B) =

11107; (C) and (D) = Pig 11212; (E) and (F) = 12509; (G) and (H) = 11907. RIN = RNA Integrity

Number.

RIN = 4.9

RIN = 2.3

RIN = 5.0

RIN = 6.8

Page 103: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

93

(A)

(B)

(C)

(D)

(E)

(F)

(G)

(H)

Figure A.4. Bioanalyzer results for normal fibrosa samples. RNA (A,C,E,G) and cDNA (B,D,F,H)

profiles indicate suitable samples for downstream microarray analysis. Pig Identifiers: (A) and (B) =

11107; (C) and (D) = Pig 11212; (E) and (F) = 12509; (G) and (H) = 11907. RIN = RNA Integrity

Number.

RIN = 6.1

RIN = 5.7

RIN = 3.6

RIN = 5.1

Page 104: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

94

A.6 Quality control plots for microarray analyses

(A)

(B)

Figure A.5. (A) Boxplot of unprocessed log-intensity distributions and (B) histogram of the density of log-

intensities between arrays. Individual arrays were labeled numerically for the bottom of lesion, non-lesion areas,

and top of lesion, respectively, for the samples 11107 (1-3), 11212 (4-6), 11907 (7-9), and 12509 (10-12).

(A)

(B)

Figure A.6. (A) Relative log expression value and (B) normalized unscaled standard error value comparisons

between arrays, which were labeled numerically for the bottom of lesion, non-lesion areas, and top of lesion,

respectively, for the samples 11107 (1-3), 11212 (4-6), 11907 (7-9), and 12509 (10-12).

11

log in

tensity

array

array

norm

aliz

ed u

nscale

d e

rror

rela

tive lo

g e

xpre

ssio

n

array

Page 105: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

95

(A)

(B)

(C)

(D)

(E)

(F)

(G)

(H)

(I)

(J)

(K)

(L)

Figure A.7. 2D images of raw probe intensity measurement for each array. Bottom of lesion, non-lesion, and top of

lesion arrays, respectively, correspond to (A-C) for sample 11107, (D-F) for sample 11212, (G-I) for sample 11907, and

(J-L) for sample 12509.

Page 106: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

96

Figure A.8. Coloured correlation coefficient matrix. The color key indicates correlation between array

intensities, which are labeled numerically for the bottom of lesion, non-lesion areas, and top of lesion,

respectively, for the samples 11107 (1-3), 11212 (4-6), 11907 (7-9), and 12509 (10-12).

Page 107: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

97

A.7 Hierarchical clustering of repeated-measures ANOVA results

12

50

9T

11

10

7T

11

21

2T

11

90

7T

11

90

7F

12

50

9F

11

90

7B

12

50

9B

11

21

2B

11

21

2F

11

10

7B

111

07F

Figure A.5. Hierarchical clustergram. One thousand forty-six transcripts identified by repeated measures

ANOVA (uncorrected p-value<0.05) as being significantly regulated among the three valve regions of interest

were clustered using unsupervised clustering analysis. A Pearson-centred correlation was used as a distance

metric with average linkage rules. Pig samples are categorized by their numerical identifiers and valve regions

are indicated by T=top of lesion, B=bottom of lesion, and F=non-lesion fibrosa. The relative expression for

each sample at a given probe is reflected by its color intensity (green=downregulated, red=upregulated).

Valve area

Sample ID

Page 108: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

98

B. Protocols

B.1 Valve leaflet histological processing for paraffin-embedded

leaflets

Purpose: To prepare FFPE samples for histological and immunohistological processing

Materials:

Ethanol (standard lab grade)

Xylene

dH2O

10% neutral buffered formalin

Water bath

Tweezers

Paraffin-embedding station

20 mL scintillation vials

Microtome

Embedding cassettes (M506-2; Simport,

Beloeil, QC, Canada)

Tissue Path disposable base molds

(22038217; 15x5x5; Fisher Scientific)

TBS Poly/FIN H-PF Paraffin

Slides (12-550-15; Fisherbrand

Superfrost Plus)

Microtome blades (819-LP; Leica

Microsystems, Concord, ON, Canada)

Slide warmer

Procedure:

Fixation

1. Place leaflet in 10% neutral buffered formalin (NBF) for 48 hrs at room temperature (10x

volume of tissue) in a scintillation vial

2. Pour off NBF and rinse twice in 70% ethanol (ETOH)

3. Fill vial with 70% ETOH and keep at 4°C until embedding

*any segmentation of the sample prior to embedding is done at this point

Dehydration and Infiltration

4. 95% ETOH 2x 30 min

5. 100% ETOH 2x 30 min

6. Xylene 2x 1 hr

7. Fill vial with paraffin wax 2x 1 hr (at 60°C)

a. Leave vial lid loose and place samples in last paraffin change into fume hood at

room temperature overnight to allow the wax to solidify and any excess xylene to

evaporate. This is done if there are many samples for embedding

Page 109: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

99

Embedding

8. Melt samples in batches in a 60°C water bath just prior to embedding

9. Dispense a little wax into the base mold and position sample with cut face touching the

base of the mold in the desired orientation using hot tweezers.

10. Touch the mold to cold surface and allow the wax to cool and slightly thicken to hold the

sample in place. Do not allow wax to totally solidify or fracture plane will occur between

wax layers.

11. Place cassette over mold and fill with wax.

12. Place on cold plate until wax is solidified.

13. Remove wax from mold using a scalpel to carefully release the sample. Samples can be

placed at -20°C prior to this to shrink the wax and help release the sample.

14. Store samples at 4°C.

Sectioning

15. Pre-heat water bath filled with ddH2O at 46°C (with no bubbles) and slide warmer at

40°C.

16. Take sample from fridge and place in microtome.

17. Cut 5 µm thick ribbons of section and transfer to clean, flat surface.

18. Cut ribbon into sets of two sections and use histology marker to number each set,

allowing multiple sets to be placed in the water bath.

19. Transfer sections to 30% ETOH room temperature bath for 1-2 min

20. Use a slide to lift samples out and slowly place into water bath. The ETOH treatment

causes the sample to stretch out when it contacts the water.

21. When sample is fully stretched out, slowly lift a side up under the section so that sections

adhere to the slide.

22. Gently flick the slide or use a Kimwipe to dry the slide and remove the majority of water

from under the section and place slide on slide warmer for 24-48 hrs or in a 40°C oven to

evaporate all water.

23. Store slides at 4°C until staining.

Page 110: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

100

B.2 Movat’s pentachrome staining for formalin-fixed, paraffin-

embedded sections

Purpose: To identify extracellular matrix components of FFPE valve tissue

Materials:

Alcian blue 1% (EMS 26385-01)

Alkaline Alcohol (EMS 26385-02)

Orcein, 0.2% (EMS 26385-03)

Hematoxylin Alcoholic, 5% (EMS

26385-04)

Ferric Chloride, 10% (EMS 26385-05)

Lugol’s Iodine (EMS 26385-06)

Woodstain Scarlet-Acid Fuchsin

Working Solution (EMS 26385-07)

Acetic Acid, 0.5% (EMS 26385-08)

Phosphotungstic Acid, 5% (EMS

26385-09)

Alcoholic Saffron, 6% (EMS 26385-10)

Bouin’s Solution (Sigma HT10132)

Ethanol

Staining dishes and racks (EMS 0312-

20)

*EMS = Electron Microscopy Sciences

Procedure: *all solutions at room temperature unless otherwise stated

1. Xylene 3x 3 min

2. 100% ETOH 3x 2 min

3. 95% ETOH 2x 2 min

4. 70% ETOH1 2x 2 min

5. 50% ETOH 2x 2 min

6. dH2O 2x 2 min

7. Bouin’s Solution in 50°C water bath – mordants tissue

*important to wash well after to remove picric acid deposits

1 hr

8. dH2O 2x 5 dips

9. Running tap water 10 min

10. dH2O dip

11. Alcian Blue 1% - stains mucosubstances blue 25 min

12. dH2O 2x 5 dips

13. Alkaline Alcohol in 56°C water bath – converts alcian blue to

monastral fast blue, which is insoluble

10 min

14. Running tap water 10 min

15. dH2O dip

16. Orcein-Verhoeff Working Solution – stains elastic fibers and nuclei 2 hr

Page 111: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

101

black **protect from light**

Immediately before use, combine in order:

1) Orcein 0.2% - 125 mL

2) Alcoholic Hematoxylin 5% - 40 mL

3) Ferric Chloride 10% - 25 mL

4) Lugol’s Iodine – 25 mL

17. dH2O 2x 5 dips

18. Ferric Chloride 2% - differentiate in solution until the elastic fibers

contrast sharply with the background **protect from light**

50 mL 10% ferric chloride + 200 mL dH2O

1.5 min

19. Running tap water 3 min

20. dH2O dip

21. Woodstain Scarlet-Acid Fuschin – stains fibrin intense red and

muscle red; cytoplasm, collagen, and ground substances will all be

red after this step

1 min

22. dH2O 2x 5 dips

23. Acetic Acid 0.5% 30 sec

24. Phosphotungstic Acid 5% - well-differentiated sections demonstrate

colorless collagen and blue-green mucopolysaccharides; it removes

red stain from collagen and ground substance

20 min

25. Acetic Acid 0.5% 30 sec

26. 100% ETOH 3x 1 min

27. Alcoholic Saffron – stains collagen and reticular fibers yellow 8 min

28. 100% ETOH 3x 2 min

29. Xylene 3x 3 min

30. Mount with non-aqueous mounting media

Results:

Nuclei = black

Cytoplasm = red

Elastic fibers = purple/black

Collagen/bone = yellow

Mucopolysaccharides = blue/green

Muscle = red

Page 112: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

102

B.3 (Immuno)histochemistry for formalin-fixed, paraffin-

embedded sections

Purpose: To determine the presence, extent, and pattern of proteoglycans/glycosaminoglycan,

Sox9, and ApoB within FFPE valve leaflets.

Materials:

PBS (-/-) diluted to 1x in ddH2O

Ethanol (histological grade)

ddH2O

Xylene

Triton (Sigma: T8532)

Water bath

Trypsin CaCl2 (Sigma: T7168)

Tween (Sigma: P1379)

Methanol

Hydrogen Peroxide

Oven

Humidity Chamber

PAP Pen

Cover slips

VectaStain Universal/Standard Elite

ABC Kit (Vector Labs: PK-6200/PK-

6100)

Vector NovaRED (Vector Labs: SK-

4800)

Hematoxylin (Vector Hematoxylin QS,

H-3404)

Synthetic mounting media (Harleco

Krystalon; 64969-71; EMD Millipore,

Billerica, MA)

Antibodies

Primary antibodies

Polyclonal sheep anti-Apolipoprotein B (ABR (Cedarlane): AHP214, 2.15 mg/mL)

Polyclonal rabbit anti-Sox9 (Abcam: ab26414, 0.6 mg/mL)

Polyclonal goat anti-biglycan (Santa Cruz: sc-27936, 0.2 mg/mL)

Polyclonal rabbit anti-decorin (Santa Cruz: sc-22753, 0.2 mg/mL)

Polyclonal rabbit anti-versican (Novus Biologicals: 16770002, 1 mg/mL)

Secondary antibodies

Biotinylated horse anti-mouse/rabbit (Universal ABC kit, Vector Labs: PK-6200)

Biotinylated rabbit anti-goat (Vector Labs: BA-5000)

Biotinylated rabbit anti-sheep (Vector Labs: BA-6000)

Biotinylated goat anti-rabbit (Vector Labs: BA-1000)

Page 113: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

103

Blocking sera

Horse serum (Universal ABC kit, Vector Labs: PK-6200)

Rabbit serum (Sigma: R9133), heat inactivated at 56°C for 30 min

Goat serum (Sigma: G9023), heat inactivated at 56°C for 30 min

IgG negative controls

Normal rabbit IgG (R&D Systems: AB-105-C, 1 mg/mL)

Normal sheep IgG (Santa Cruz: sc-2717)

Normal goat IgG (R&D Systems: AB-108-C, 1 mg/mL)

Table B.1. Immunohistochemistry summary

Primary Antibody Vector Labs Kit Antigen Retrieval Secondary

Antibody

Control Tissue

Antibody Concentration

ApoB 3.6 µg/mL Standard Trypsin-CaCl2 anti-sheep Bone

Sox9 3 µg/mL Standard Tris EDTA anti-rabbit Cartilage

Biglycan 20 µg/mL Standard Trypsin-CaCl2 anti-goat Heart Valve

Decorin 10 µg/mL Universal Trypsin-CaCl2 anti-rabbit Heart Valve

Versican 1.3 µg/mL Universal Trypsin-CaCl2 anti-rabbit Heart Valve

Protocol for Standard Immunohistochemistry:

1. Bake slides, 30 min, 60°C oven

2. Dewax/rehydration

a. Xylene 3x 5 min

b. 100% ETOH 3x 5 min

c. 95% ETOH 1x 5 min

d. 70% 1x 5 min

e. ddH2O 1x 5 min

3. Antigen retrieval

a. Enzymatic antigen retrieval: 120µL Trypsin-CaCl2 (1mg/ml) + 120mL ddH2O at

37°C in water bath for 30 min

b. Heat mediated antigen retrieval: Tris-EDTA Buffer (10 mM Tris, 1 mM EDTA,

0.05% Tween, pH 9.0) at 98°C in water bath for 20 min, then cooled in room

temperature water for 20 min, and rinsed in ddH2O

4. Wash – PBS/0.05% Tween 3x 3 min

5. Peroxidase block – 3% H2O2/Methanol (10.5mL 30% H2O2 + 94.5mL methanol) for 10

min at room temperature

Page 114: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

104

6. Wash – PBS/0.05% Tween 2x 3 min

7. Keep sections hydrated with PBS, while outlining sections with a PAP pen

8. Serum block at room temperature in humidity chamber for 45 min

a. 1 drop (Universal kit = 50µL or Standard kit = 75 µL) of appropriate stock serum

+ 5mL PBS

9. Primary antibody at room temperature in humidity chamber for 1 hr

a. Antibody is diluted to appropriate concentration in 0.3% TritonX-100 in PBS

b. 0.3% TritonX-100: 3 µL Triton + 1 mL PBS

c. Only Triton/PBS or IgG of primary on negative controls

10. Wash – PBS/0.05% Tween 4x 3 min

11. Secondary antibody at room temperature in dark humidity chamber for 30 min

a. 1 drop (Universal kit = 50µL or Standard kit = 75 µL) of appropriate stock serum

+ 2.5mL PBS + 1 drop (Universal kit = 50 µL or Standard kit = 12.5 µL) of

appropriate biotinylated antibody

12. Wash – PBS/0.05% Tween 4x 3 min

13. VectaStain ABC at room temperature in dark humidity chamber for 30 min

a. 1 drop Reagent A + 2.5 mL PBS + 1 drop Reagent B (allow to stand for 30 min at

room temperature in the dark before use)

14. Wash – PBS/0.05% Tween 2x 3 min

15. Vector NovaRED at room temperature in the dark for 5 min

a. 5 mL dH2O

b. 50 µL Reagent #1 mix well

c. 50 µL Reagent #2 mix well

d. 45 µL Reagent #3 mix well

e. 75 µL hydrogen peroxide solution mix well

16. Wash – dH2O 1x 3 min

17. Rinse slides in tap water

18. Counterstain with Vector Hematoxylin QS on slide for 10 sec

a. Rinse hematoxylin off slides with tap water (until water becomes colourless)

19. Dehydration

a. 70% ETOH 1x 5 min

b. 95% ETOH 1x 5 min

c. 100% ETOH 2x 5 min

Page 115: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

105

d. Xylene 2x 5 min

20. Mount coverslips onto slides with synthetic mounting media

Protocol for Hyaluronan Staining:

Additional Materials

Hyaluronidase from Streptomyces hyalurolyticus (Sigma: H1136, 882 U/vial)

o Reconstitute in 4mL of 20 mM sodium phosphate buffer (77 mM sodium

chloride, 0.01% BSA, pH 7.0) – resulting concentration: 220.5 U/mL

Hyaluronan-binding protein (Calbiochem: 385911, 500 µg/mL)

Control Slide Preparation

1. Bake slides at 60°C for 30 min

2. Dewax/rehydration

a. Xylene 3x 5 min

b. 100% ETOH 3x 5 min

c. 95% ETOH 1x 5 min

d. 70% 1x 5 min

e. ddH2O 1x 5 min

3. Treat slides with hyaluronidase in PBS – 100 U/mL at 37°C for 4 hrs

4. Store slides at 4°C in PBS until use (*add into slide set at peroxidase block – step 3)

Histochemistry Protocol

1. Bake slides, 30 min, 60°C oven

2. Dewax/rehydration

a. Xylene 3x 5 min

b. 100% ETOH 3x 5 min

c. 95% ETOH 1x 5 min

d. 70% 1x 5 min

e. ddH2O 1x 5 min

3. Peroxidase block – 3% H2O2/Methanol (10.5mL 30% H2O2 + 94.5mL methanol) for 10

min at room temperature

4. Wash – PBS/0.05% Tween 2x 3 min

5. Keep sections hydrated with PBS, while outlining sections with a PAP pen

6. 3% BSA block at room temperature for 45 min

Page 116: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

106

7. Incubate with 110.25 U/mL biotinylated hyaluronan-binding protein at room temperature

in humidity chamber for 1 hr

a. 250 µL biotinylated hyaluronan-binding protein (220.5 U/mL) + 250 µL PBS

8. Wash – PBS/0.05% Tween 4x 3 min

9. VectaStain ABC at room temperature in dark humidity chamber for 30 min

a. 1 drop Reagent A + 2.5 mL PBS + 1 drop Reagent B (allow to stand for 30 min at

room temperature in the dark before use)

10. Wash – PBS/0.05% Tween 2x 3 min

11. Vector NovaRED at room temperature in the dark for 5 min

a. 5 mL dH2O

b. 50 µL Reagent #1 mix well

c. 50 µL Reagent #2 mix well

d. 45 µL Reagent #3 mix well

e. 75 µL hydrogen peroxide solution mix well

12. Wash – dH2O 1x 5 min

13. Rinse slides in tap water

14. Counterstain with Vector Hematoxylin QS on slide for 10 sec

a. Rinse hematoxylin off slides with tap water (until water becomes colourless)

15. Dehydration

a. 70% ETOH 1x 5 min

b. 95% ETOH 1x 5 min

c. 100% ETOH 2x 5 min

d. Xylene 2x 5 min

16. Mount coverslips onto slides with synthetic mounting media

B.4 Image processing

Purpose: To separate proteoglycan (PG) staining areas from Nova Vector Red (NVR) images

taken using the Aperio ScanScope XT Scanner at the Princess Margaret Hospital (PMH)

Advanced Optical Microscope Facility.

Materials:

ImageJ (version 1.47v)

Page 117: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

107

G. Landini’s Colour Thresholding v1.9 plugin

ImageJ Calculator Plus plugin

Colour Separation:

//Thresholds for use with NVR PG images taken at PMH

//Adapted from macros created by Krista Sider

dir1 = getDirectory("Choose SOURCE Directory ");

dir2 = getDirectory("Choose MASK Directory ");

dir3 = getDirectory("Choose INVERSE mask Directory ");

list = getFileList(dir1);

setBatchMode(true);

format = "Measurements"

format2 = "8-bit TIFF"

run("Set Measurements...", "area limit display redirect=None decimal=3");

run("Options...", "iterations=1 black edm=Overwrite count=1");

run("Colors...", "foreground=white background=black selection=yellow");

run("Options...", "iterations=1 black count=1");

//Open Image to set scale then close - when commented out does measurement in pixels

open(dir1+list[0]);

//Removes Scale

run("Set Scale...", "distance=0 known=0 pixel=1 unit=pixel global");

close();

//Runs NVR Separation 1

for (j=0; j<list.length; j++) {

//Open Image

open(dir1+list[j]);

titleSample = getTitle;

//gets length of title and removes ".tif" from the file name

newname = substring(titleSample, 0, lengthOf(titleSample)-4);

run("Threshold Colour");

// Colour Thresholding v1.9-------

// Autogenerated macro, single images only!

// G Landini 2/Feb/2008.

//

// This only works with Black background and White foreground

min=newArray(3);

Page 118: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

108

max=newArray(3);

filter=newArray(3);

a=getTitle();

run("HSB Stack");

run("Convert Stack to Images");

selectWindow("Hue");

rename("0");

selectWindow("Saturation");

rename("1");

selectWindow("Brightness");

rename("2");

//Hue Values

min[0]=35;

max[0]=220;

filter[0]="stop";

//Saturation Values

min[1]=15;

max[1]=255;

Mask Area Measurement:

//Created Jan 2012 by Krista Sider

dir1 = getDirectory("Choose SOURCE Mask Directory ");

dir2 = getDirectory("Choose RESULTS Directory ");

list = getFileList(dir1);

setBatchMode(true);

format = "Measurements"

run("Set Measurements...", "area limit display redirect=None decimal=3"); //measures area

run("Options...", "iterations=1 black edm=Overwrite count=1");

//Open Image to set scale then close

open(dir1+list[0]);

//Removes Scale

run("Set Scale...", "distance=0 known=0 pixel=1 unit=pixel global");

close();

// Measures the NVR Mask Area

for (i=0; i<list.length; i++) {

Page 119: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

109

//Open Masked Image

open(dir1+list[i]);

//Convert to 8-bit image so can measure

run("8-bit");

//Make a Threshold for Measurement

setAutoThreshold();

//Measure Threshold Area

run("Measure");

close();

}

B.5 Valve leaflet histological processing for OCT-embedded

leaflets

Purpose: To prepare frozen, OCT-embedded samples for histological and immunohistological

processing

Materials:

RNaseZap and DNA Zap

RNase/DNase-free H2O

RNase-free PBS

Petri dishes

Moulds

OCT (VWR: 25608-930)

Kimwipes

Dry ice

Scissors

Tweezers

Liquid nitrogen

Ziploc bag

Protocol: **ensure all surfaces, solutions, and equipment are RNase/DNA-free

OCT-Embedding and Cryopreservation

1. Pour RNase-free PBS into petri dishes.

2. Fill bottom of mould with OCT.

3. Rinse leaflet and syringe needle in RNase-free PBS in one of the petri dishes.

a. The side of the leaflet facing the hub of the needle = aortic side

b. The side of the leaflet facing the tip of the needle = ventricular side

Page 120: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

110

4. Remove the leaflet from the needle onto a dry petri dish, ensuring they are in the same

orientation. Cut the leaflet in half in the radial direction with scissors.

5. Rinse each leaflet half in PBS again, leaving them in PBS until they are ready to be

placed in the mould.

6. On a dry petri dish, lightly blot one leaflet half dry with Kimwipes.

7. Place that leaflet half in the mould partly filled with OCT with the aortic side up and the

midline towards the end of the mould without extra plastic.

a. Align the leaflet so it is flat, straight and aligned parallel to the edge of the mould.

b. Leave some room between the midline side of the leaflet and the edge of the

mould to allow for blade alignment during cryosectioning.

8. Top up the mould with OCT until it forms a small meniscus.

9. Place a labelled paper tag into the side with extra plastic.

10. Touch the bottom of the mould to liquid nitrogen for at most 60 seconds.

a. Just lower the mould until you can hear the liquid nitrogen boiling.

11. Once the OCT is completely white and hard (check the centre of the mould), remove

from liquid nitrogen.

12. If the OCT has bubbled and/or the valve tissue is still exposed on top of the mould, add

more OCT into the space that has formed and again, touch the bottom of the mould to

liquid nitrogen until the new OCT is white and hard.

13. Place mould with leaflet in a sealed Ziploc bag directly on dry ice for temporary storage.

14. Store leaflets at -80°C long term.

B.6 Movat’s pentachrome staining for frozen OCT-embedded

sections

Purpose: To identify extracellular matrix components of frozen OCT-embedded valve tissue

Materials:

Alcian blue 1% (EMS 26385-01)

Alkaline Alcohol (EMS 26385-02)

Orcein, 0.2% (EMS 26385-03)

Hematoxylin Alcoholic, 5% (EMS

26385-04)

Ferric Chloride, 10% (EMS 26385-05)

Lugol’s Iodine (EMS 26385-06)

Woodstain Scarlet-Acid Fuchsin

Working Solution (EMS 26385-07)

Acetic Acid, 0.5% (EMS 26385-08)

Phosphotungstic Acid, 5% (EMS

26385-09)

Page 121: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

111

Alcoholic Saffron, 6% (EMS 26385-10)

Bouin’s Solution (Sigma HT10132)

Ethanol

Staining dishes and racks (EMS 0312-

20)

*EMS = Electron Microscopy Sciences

Procedure: *all solutions at room temperature unless otherwise stated

1. Warm slide box to room temperature before opening slides 30 min

2. Neutral buffered formalin 10 min

3. Running Tap water 3 min

4. Bouin’s Solution in 50°C water bath – mordants tissue

*important to wash well after to remove picric acid deposits

1 hr

5. dH2O 2x 5 dips

6. Running tap water 10 min

7. dH2O dip

8. Alcian Blue 1% - stains mucosubstances blue 50 min

9. dH2O 2x 5 dips

10. Alkaline Alcohol in 56°C water bath – converts alcian blue to

monastral fast blue, which is insoluble

10 min

11. Running tap water 10 min

12. dH2O dip

13. Orcein-Verhoeff Working Solution – stains elastic fibers and nuclei

black **protect from light**

Immediately before use, combine in order:

5) Orcein 0.2% - 125 mL

6) Alcoholic Hematoxylin 5% - 40 mL

7) Ferric Chloride 10% - 25 mL

8) Lugol’s Iodine – 25 mL

2 hr

14. dH2O 2x 5 dips

15. Ferric Chloride 2% - differentiate in solution until the elastic fibers

contrast sharply with the background **protect from light**

50 mL 10% ferric chloride + 200 mL dH2O

3.5 min

16. Running tap water 3 min

17. dH2O dip

18. Woodstain Scarlet-Acid Fuschin – stains fibrin intense red and

muscle red; cytoplasm, collagen, and ground substances will all be

red after this step

1 quick dip

19. dH2O 2x 5 dips

20. Acetic Acid 0.5% 30 sec

21. Phosphotungstic Acid 5% - well-differentiated sections demonstrate 30 min

Page 122: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

112

colorless collagen and blue-green mucopolysaccharides; it removes

red stain from collagen and ground substance

22. Acetic Acid 0.5% 30 sec

23. 100% ETOH 3x 1 min

24. Alcoholic Saffron – stains collagen and reticular fibers yellow 8 min

25. 100% ETOH 2x 3 min

26. Xylene 3x 3 min

27. Mount with non-aqueous mounting media

Results:

Nuclei = black

Cytoplasm = red

Elastic fibers = purple/black

Collagen/bone = yellow

Mucopolysaccharides = blue/green

Muscle = red

B.7 Immunohistochemistry staining for frozen OCT-embedded

sections

Purpose: To determine the presence, extent, and pattern of Sox9 in frozen OCT-embedded valve

leaflets.

Materials:

Acetone

PBS (-/-) diluted to 1x in ddH2O

Ethanol (histological grade)

ddH2O

Xylene

Triton (Sigma: T8532)

Water bath

Tween (Sigma: P1379)

Methanol

Hydrogen Peroxide

Oven

Humidity Chamber

PAP Pen

Cover slips

VectaStain Standard Elite ABC Kit

(Vector Labs: PK-6100)

Vector NovaRED (Vector Labs: SK-

4800)

Hematoxylin (Vector Hematoxylin QS,

H-3404)

Synthetic mounting media (Harleco

Krystalon; 64969-71; EMD Millipore,

Billerica, MA)

Page 123: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

113

Antibodies and Serum

Polyclonal rabbit anti-Sox9 (Abcam: ab26414, 0.6 mg/mL)

Biotinylated goat anti-rabbit (Vector Labs: BA-1000)

Goat serum (Sigma: G9023), heat inactivated at 56°C for 30 min

Normal rabbit IgG (R&D Systems: AB-105-C, 1 mg/mL)

Protocol:

1. Warm slide box at room temperature for 30 min

a. During this time, pre-cool acetone at -20°C

2. Fixation in pre-cooled acetone at room temperature for 10 min

3. Wash – PBS/0.05% Tween 3x 3 min

4. Peroxidase block – 3% H2O2/Methanol (10.5mL 30% H2O2 + 94.5mL methanol) for 10

min at room temperature

5. Wash – PBS/0.05% Tween 2x 3 min

6. Keep sections hydrated with PBS, while outlining sections with a PAP pen

7. Serum block at room temperature in humidity chamber for 45 min

a. 1 drop (Standard kit = 75 µL) of goat serum + 5mL PBS

8. Primary antibody at room temperature in humidity chamber for 1 hr

a. Antibody is diluted to appropriate concentration in 0.3% TritonX-100 in PBS

b. 0.3% TritonX-100: 3 µL Triton + 1 mL PBS

c. Only Triton/PBS or IgG of primary on negative controls

9. Wash – PBS/0.05% Tween 4x 3 min

10. Secondary antibody at room temperature in dark humidity chamber for 30 min

a. 1 drop (Standard kit = 75 µL) of goat serum + 2.5mL PBS + 1 drop (Standard kit

= 12.5 µL) of biotinylated goat anti-rabbit secondary

11. Wash – PBS/0.05% Tween 4x 3 min

12. VectaStain ABC at room temperature in dark humidity chamber for 30 min

a. 1 drop Reagent A + 2.5 mL PBS + 1 drop Reagent B (allow to stand for 30 min at

room temperature in the dark before use)

13. Wash – PBS/0.05% Tween 2x 3 min

14. Vector NovaRED at room temperature in the dark for 5 min

a. 5 mL dH2O

Page 124: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

114

b. 50 µL Reagent #1 mix well

c. 50 µL Reagent #2 mix well

d. 45 µL Reagent #3 mix well

e. 75 µL hydrogen peroxide solution mix well

15. Wash – dH2O 1x 5 min

16. Rinse slides in tap water

17. Counterstain with Vector Hematoxylin QS on slide for 10 sec

a. Rinse hematoxylin off slides with tap water (until water becomes colourless)

18. Dehydration

a. 70% ETOH 1x 5 min

b. 95% ETOH 1x 5 min

c. 100% ETOH 2x 5 min

d. Xylene 2x 5 min

19. Mount coverslips onto slides with synthetic mounting media

B.8 Laser capture microdissection

Purpose: To capture cells from areas of interest in frozen OCT-embedded valve leaflet sections.

**ensure all surfaces, solutions, and equipment are RNase/DNA-free

Tissue Staining:

Materials

Arcturus Histogene staining solution

(Life Technologies, KIT0415)

ETOH

Xylene

dH2O (Life Technologies, 10977015)

50 mL conical tubes

Tweezers

Preparation

Set out 9 conical tubes in the following order (with 25 mL solution/tube):

1) 70% ETOH (1)

2) dH2O (1)

3) dH2O (2)

4) 70% ETOH (2)

5) 95% ETOH

6) 100% ETOH (1)

7) 100% ETOH (2)

8) Xylene (1)

Page 125: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

115

9) Xylene (2)

Set out two sets of Kimwipes: (1) to blot and (2) to air dry xylene

Loosen caps only up to 95% ETOH wash to prevent H2O from getting into 100% ETOH

Preload pipette with 100 µL HistoStain before starting protocol

Protocol *one slide at a time*

1. Fix in 70% ETOH (1) for 30 sec

2. Rinse in dH2O (1) for 30 sec – shake vigorously to get rid of OCT

a. Blot, flick, and wipe slide before next step

3. Stain with 100 µL HistoStain for 20 sec – don’t shake bottle before preloading pipette

a. Take off stain with pipette (can reuse for 2-3 slides)

4. Rinse in dH2O (2) for 30 sec

a. Blot, flick, and wipe slide before next step

5. Dehydrate

a. 70% ETOH (2) for 30 sec

b. 95% ETOH for 30 sec

c. 100% ETOH (1) for 1 min

d. 100% ETOH (2) for 1 min

6. Xylene (1) for 5 min can start next slide after current slide has been placed into this

wash *wipe tweezers before transfer to Xylene (2)

7. Xylene (2) for 5 min

8. Air dry for 10-20 min

Laser Capture Microdissection:

Materials

Arcturus PixCell II Laser Capture

System

Arcturus CapSure Macro LCM Caps

(Life Technologies, LCM0211)

Arcturus PicoPure RNA Isolation kit

(Life Technologies, KIT0204)

500 µL Eppendorf tubes

Sticky notes

Tweezers

1% SDS

70% ETOH

Blade

Thermocycler

Page 126: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

116

Preparation

1) Turning on the microscope:

a. Wipe down microscope with 1% SDS and rinse with 70% ETOH

b. Turn ON: (1) Scope system – with light intensity set to lowest setting

(2) Computer

(3) Software (ver 2.0.0)

*view screen should be black; adjust light to test that program is not frozen;

should be at “Norm” setting on start-up with knob turned fully counter-clockwise

**if frozen, turn off everything in reverse order and restart

c. Turn the key ¼-turn clockwise to actuate the laser

d. After laser interlock has been checked for continuity, turn on laser with the

“ENABLE’ button

2) Loading the cap

a. Load Arcturus CapSure Macro LCM Caps

i. Push knobs in to unlock

ii. Push loader down and slide cap cassette to far end at the “LOAD” line on

the stage

iii. Pull knobs out on both sides of the loader to lock caps in place

b. Swing the capping arm counter-clockwise until it stops over the new cap

c. Raise the capping arm to detach the new cap from the cassette and move it

clockwise to the rest position

3) Focusing the laser

a. Position joystick so it is perpendicular to the tabletop

b. Place the slide into position so that it is centered on the desired capture area and

the slide is above the H-groove

c. Turn ON vacuum to secure slide position and focus the stage on the tissue

d. Always focus the laser at the smallest spot size

e. Fire test pulse on area away from tissue

i. Power = increase to get appropriate burn spot

ii. Duration = increase to adhere to tissue

**a focused laser should have a uniformly, black thick circle of melted

plastic on the cap with a black ring

Page 127: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

117

4) Capturing cells

a. Position cells of interest under laser

b. Fire the laser on the cells, using the joystick to adjust the position of the tissue

relative to the stationary laser

c. When enough cells have been collected, raise the capping arm and move it to the

rest position

d. Observe the holes left behind in the tissue

e. To observe the captured material:

i. Release the vacuum

ii. Move the slide so that there is clear glass over the objective view

iii. Turn ON the vacuum

iv. Return the capping arm to the work position

f. Repeat until sufficient cells have been captured on the cap

5) Removing the Cap

a. Move the arm to the capping station to drop up the cap (fully up, fully counter-

clockwise, then lower)

b. Move the arm clockwise without lifting to expose the completed cap

c. Use the capping tool to lift the cap

d. Use the adhesive from a new sheet of sticky note to remove unwanted tissue from

the cap

i. View the cap under the microscope to ensure all unwanted tissue is

removed

e. Use a blade to cut off excess areas of the cap that do not contain tissue

f. Use tweezers to peel transfer film off cap and place in a 500 µL Eppendorf tube

6) RNA extraction

a. Pipette 50 µL Extraction Buffer into the 500 µL Eppendorf tube containing

isolated tissue

b. Incubate assembly for 30 min at 42°C in Thermocycler

c. Freeze cell extract at -80°C until isolation

Page 128: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

118

B.9 RNA isolation

Purpose: To isolate RNA from laser captured valve sections.

**ensure all surfaces, solutions, and equipment are RNase/DNA free

Materials:

Arcturus PicoPure RNA Isolation kit (Life Technologies: KIT0204)

RNase-free DNase I (Qiagen: 79254)

Centrifuge

Protocol:

1) Pipette 250 µL Conditioning Buffer onto purification column

a. Incubate for 5 min at room temperature

b. Centrifuge at 16,000 x g for 1 min

2) Pipette 50 µL ETOH into cell extract from RNA extraction

a. Pipette up and down to mix well

3) Pipette cell extract and ETOH mixture into purification column

a. Centrifuge at 100 x g for 2 min binds RNA to column

b. Centrifuge at 16,000 x g for 30 sec removes flowthrough

4) Pipette 100 µL Wash Buffer 1 into column

a. Centrifuge at 8,000 x g for 1 min

5) DNase treatment:

a. 5 µL DNase I Stock Solution + 35 µL Buffer RDD – invert gently to mix

b. Pipette the 40 µL DNase mix into column

c. Incubate for 15 min at room temperature

d. Pipette 40 µL Wash Buffer 1 into column

e. Centrifuge at 8,000 x g for 15 sec

6) Pipette 100 µL Wash Buffer 2 into column

a. Centrifuge at 8,000 x g for 1 min

7) Pipettte another 100 µL Wash Buffer 2 into column

a. Centrifuge at 16,000 x g for 2 min

b. If there is residual wash buffer, re-centrifuge at 16,000 x g for 1 min

Page 129: Characterization of Changes in Extracellular Matrix and ... · Andrea Victoria Kwong Master of Applied Science in Biomedical Engineering Institute of Biomaterials and Biomedical Engineering

119

8) Transfer column to new 0.5 mL tube from the kit

9) Pipette 11 µL Elution Buffer directly onto membrane of column (gently touch)

a. Incubate column for 1 min at room temperature

b. Centrifuge column at 1,000 x g for 1 min distributes Elution Buffer in column

c. Centrifuge at 16,000 x g for 1 min elutes RNA

10) Store sample at -80°C until use