mechanisms and reversal of elastin specific medial

214
Clemson University TigerPrints All Dissertations Dissertations 8-2014 MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL ARTERIAL CALCIFICATION Yang Lei Clemson University, [email protected] Follow this and additional works at: hps://tigerprints.clemson.edu/all_dissertations is Dissertation is brought to you for free and open access by the Dissertations at TigerPrints. It has been accepted for inclusion in All Dissertations by an authorized administrator of TigerPrints. For more information, please contact [email protected]. Recommended Citation Lei, Yang, "MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL ARTERIAL CALCIFICATION" (2014). All Dissertations. 1307. hps://tigerprints.clemson.edu/all_dissertations/1307

Upload: others

Post on 07-Dec-2021

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

Clemson UniversityTigerPrints

All Dissertations Dissertations

8-2014

MECHANISMS AND REVERSAL OFELASTIN SPECIFIC MEDIAL ARTERIALCALCIFICATIONYang LeiClemson University, [email protected]

Follow this and additional works at: https://tigerprints.clemson.edu/all_dissertations

This Dissertation is brought to you for free and open access by the Dissertations at TigerPrints. It has been accepted for inclusion in All Dissertations byan authorized administrator of TigerPrints. For more information, please contact [email protected].

Recommended CitationLei, Yang, "MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL ARTERIAL CALCIFICATION" (2014). AllDissertations. 1307.https://tigerprints.clemson.edu/all_dissertations/1307

Page 2: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

ARTERIAL CALCIFICATION

A Dissertation

Presented to the

Graduate School of

Clemson University

In Partial Fulfillment

of the Requirements for the Degree

Doctor of Philosophy

Bioengineering

byYang Lei

August 2014

Accepted by: Dr. Narendra Vyavahare, Committee Chair

Dr. Jeoung Soo Lee Dr. Alexey Vertegel

Dr. Bruce Z. Gao

Page 3: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

II

ABSTRACT

There are two distinctive types of vascular calcification: atherosclerotic intimal

calcification (AIC) and elastin specific medial arterial calcification (MAC). AIC occurs

in the context of atherosclerosis, associated with lipids, macrophages, and vascular

smooth muscle cells; whereas, MAC can exist independently of atherosclerosis and is

associated with elastin and vascular smooth muscle cells. The work presented here

focuses on the mechanisms and reversal of MAC. MAC, termed as Monckeberg’s

sclerosis, is a type of vascular calcification disease mostly occurs as linear deposits along

elastic lamellae. Vascular calcification, including medial calcification, is a strong

predictor of cardiovascular morbidity and mortality. Currently the only options to treat

vascular calcifications are surgical methods like directional atherectomy and placement

of stent grafts. These surgical methods are quite invasive and expensive, and currently

there is no available alternative to surgical therapy for reversal of vascular calcification.

One possible approach is EDTA chelation therapy. EDTA chelation therapy most often

involves the injection of disodium ethylene diamine tetraacetic acid (EDTA), a chemical

that binds, or chelate ionic calcium, trace elements and other divalent cations so as to

reverse calcification. However, chelation therapy is not yet accepted in US and not

approved by FDA as there is no clinical proof that it works and improves cardiovascular

function. Besides, it has severe side effects such as renal toxicity, hypercalcemia, and

bone loss.

Page 4: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

III

Overall goal of our research is to understand the mechanisms of elastin specific

MAC, to evaluate efficacy of chelating agents to reverse elastin specific MAC, and to

develop a targeted delivery system to reverse elastin specific MAC and circumvent side

effects.

First, we investigated the mechanisms of elastin specific MAC, specifically cause-

and-effect relationship between the elastin-specific MAC and osteoblast-like

differentiation of vascular smooth muscle cells (VSMCs). It has been accepted for

decades that the pathology of vascular calcification resembles physiological bone

mineralization. However, the cause-and-effect relationship between calcification and

osteoblast-like differentiation of VSMCs is still unclear. We, for the first time, showed

that in response to the calcified matrix, for both hydroxyapatite crystals and calcified

aortic elastin, VSMCs lost their smooth muscle lineage markers and underwent

chondroblast/osteoblast-like differentiation. Interestingly, this phenotypic transition was

reversible and VSMCs restored their original linage upon reversal of calcification.

Secondly, we evaluated the efficacy of chelating agents to reverse elastin specific

MAC. We tested if chelating agents, such as disodium ethylene diamine tetraacetic acid

(EDTA), diethylene triamine pentaacetic acid (DTPA) and sodium thiosulfate (STS) can

remove calcium from hydroxyapatite (HA) and calcified tissues without damaging the

tissue architecture. Our results indicated that both EDTA and DTPA could effectively

remove calcium from hydroxyapatite and calcified tissues, while STS was not effective.

The tissue architecture was not altered during chelation.

Page 5: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

IV

Thirdly, we investigated the therapeutic effects of systemic EDTA chelation

therapy for reversal of calcification and its possible side effects in vivo. We used CaCl2

mediated abdominal aorta injury in rats to mimic local aortic calcification similar to what

is observed in clinical cases. We demonstrated that systemic EDTA chelation therapy did

not reverse local aortic calcification. Moreover, systemic EDTA chelation therapy caused

adverse events for serum and urine calcium, specifically hypocalcemia and

hypercalciuria, but did not cause bone loss.

Finally, we developed a targeted delivery system to reverse elastin specific MAC

and to avoid side effects associated with systemic EDTA chelation therapy. We have

recently shown that the nanoparticles (NPs) coated with elastin-antibody can be targeted

to vascular calcification sites. This paved the way to design an elastin antibody coated

and EDTA loaded albumin NPs to reverse elastin-specific medial calcification and avoid

side effects. Our in vitro and in vivo results demonstrated that elastin antibody coated and

EDTA loaded albumin NPs had good therapeutic effect to reverse elastin specific MAC

and did not have side effects typically seen in systemic EDTA chelation therapy such as

hypocalcemia and bone loss.

In conclusion, our work led to enhanced understanding the possible mechanisms

of osteogenesis in vascular calcification and possible alternative therapy to reverse elastin

specific MAC without systemic side effects.

Page 6: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

V

DEDICATION

I would like to dedicate this work to my parents and my brother. My family is my

greatest motivation to complete this dissertation.

Page 7: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

VI

ACKNOWLEDGEMENTS

I would like to give special thanks to my advisor Dr. Naren Vyavahare for giving

me an opportunity to work with him on this project. My previous research background

during master’s and bachelor’s degree was related to materials science and engineering.

However, Dr. Vyavahare still trusted me and gave me great support to work on this

project in department of bioengineering. Dr. Vyavahare always encouraged me and was

available to answer my doubts and to give me insight into directions of research goals.

His rigorous academic attitude, friendly manner to colleagues and students set a good

example for my future academic career.

I am especially thankful to my PhD committee members Dr. Jeoung Soo Lee, Dr.

Alexey Vertegel, and Dr. Bruce Z. Gao. Dr. Jeoung Soo Lee taught me all cellular and

molecular biology analyses techniques. Dr. Alexey Vertegel helped me a lot for

nanoparticles’ techniques including preparation, characterization and optimization. Dr.

Bruce Z. Gao helped me a lot for optical imaging. I would also like to thank previous lab

member, Aditi Sinha, who always helped me with laboratory techniques and

troubleshooting.

I would also like to thank all other Cardiovascular ECM Stabilization and

Regeneration (CESR) Lab members including: Nasim Nosoudi, Pranjal Nahar, Saketh

Karamched, Vaideesh Parasaram, Hobey Tam, Linae Maganini and Aniqa Chowdhury

for their help for lab works; all administrative staff at department of Bioengineering at

Clemson University: Maria Torres, Leigh Humphries, Michelle Kirby, Sherri Morrison

Page 8: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

VII

who always kindly to provide help; Linda Jenkins and Chad L. McMahan at department

of Bioengineering, Clemson University for histology lab work; Dr. Hamp Bruner for

histology examination; Dr. John Parrish, Travis Pruitt, Tina Parker and Stefanie Pardue at

Godley-Snell Research Center (Clemson, SC) of Clemson University for animal work;

Dr. Terri Bruce and Rhonda Powell at Clemson Light Imaging Facility (Clemson, SC) of

Clemson University for optical microscope work; Dr. Lax Saraf, Dr. Haijun Qian and

Dayton Cash at Clemson University Electron Microscopy Laboratory(Anderson, SC) for

electron microscope work; Dr. David Kwartowitz, Michael Jaeggli and Siyu Ma at

Clemson University for Micro-CT data analysis using Mevislab software; Luke

Pietrykowski at the Clemson University Biomedical Engineering Innovation Campus

(Greenville, SC) for mechanical test using Bose Electroforce 3200; Dr. Michael E. Ward

at Greenville Hospital System (Greenville, SC) for providing us samples of calcified

human aorta; Dr. Jonathon Nye, Margie Jones and Dr. Baowei Fei at Center for Systems

Imaging, Emory University School of Medicine (Atlanta, GA) for Micro-CT work.

I am also grateful to the Department of Bioengineering and Clemson University

for being my home for last four years in my PhD life. Finally, I would like to

acknowledge the funding provided by National Institutes of health (P20GM103444,

R01HL061652) and the Hunter Endowment (All awarded to Dr. Naren Vyavahare).

Page 9: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

VIII

TABLE OF CONTENTS

TITLE PAGE ..................................................................................................................... I

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

DEDICATION.................................................................................................................. V

ACKNOWLEDGEMENTS ........................................................................................... VI

TABLE OF CONTENTS ............................................................................................ VIII

LIST OF TABLES ....................................................................................................... XIII

LIST OF FIGURES ..................................................................................................... XIV

CHAPTERS ....................................................................................................................... 1

CHAPTER 1: INTRODUCTION .................................................................................... 1

CHAPTER 2: LITERATURE REVIEW ....................................................................... 7

2.1 Vascular architecture ................................................................................................. 7

2.1.1 Normal anatomy of blood vessel ........................................................................... 7

2.1.2 Structure of aortic elastin ....................................................................................... 8

2.2 Vascular diseases related to calcification ................................................................ 10

Page 10: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

IX

2.2.1 Atherosclerosis ..................................................................................................... 10

2.2.2 Aortic aneurysms ................................................................................................. 12

2.3 Vascular calcification................................................................................................ 14

2.3.1 Medial arterial calcification (MAC) .................................................................... 14

2.3.2 Mechanisms of vascular calcification .................................................................. 15

2.3.3 Types and characteristics of vascular calcification .............................................. 18

2.3.4 Animal models for investigating vascular calcification ....................................... 21

2.3.5 Current or prospective therapies for treating vascular calcification .................... 27

2.4 Chelation therapy...................................................................................................... 30

2.4.1 Types and applications of chelation therapy ........................................................ 31

2.4.2 In vitro and animal studies of chelation therapy for vascular calcification ......... 34

2.5 EDTA chelation therapy........................................................................................... 37

2.5.1 Case studies and clinical trials of EDTA chelation therapy ................................ 37

2.5.2 Side effects with traditional systemic EDTA chelation therapy .......................... 41

2.5.3 Current opinions of EDTA chelation therapy for cardiovascular disease ........... 44

2.6 Targeted EDTA chelation therapy .......................................................................... 46

2.6.1 Local drug delivery in the vasculature ................................................................. 46

2.6.2 Albumin nanoparticles as drug carriers for different application ........................ 50

2.6.3 Targeted EDTA chelation therapy using EDTA loaded albumin nanoparticles .. 52

CHAPTER 3: PROJECT RATIONALE ...................................................................... 53

Page 11: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

X

3.1 Project objective and aims ....................................................................................... 53

3.2 Specific aims and rationale ...................................................................................... 53

3.3 Clinical significance .................................................................................................. 56

CHAPTER 4: MECHANISMS OF ELASTIN SPECIFIC MEDIAL ARTERIAL

CALCIFICATION .......................................................................................................... 58

4.1 Introduction ............................................................................................................... 58

4.2 Materials and methods ............................................................................................. 59

4.3 Results ........................................................................................................................ 66

4.4 Discussion................................................................................................................... 77

4.5 Conclusion ................................................................................................................. 83

CHAPTER 5: EFFICACY OF CHELATING AGENTS TO REVERSE ELASTIN

SPECIFIC MEDIAL ARTERIAL CALCIFICATION .............................................. 86

5.1 Introduction ............................................................................................................... 86

5.2 Materials and methods ............................................................................................. 87

5.3 Results ........................................................................................................................ 93

5.4 Discussion................................................................................................................. 103

Page 12: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XI

5.5 Conclusion ............................................................................................................... 108

CHAPTER 6: SYSTEMIC EDTA CHELATION THERAPY FAILED TO

REVERSE MAC AND HAD SIDE EFFECTS .......................................................... 110

6.1 Introduction ............................................................................................................. 110

6.2 Materials and methods ........................................................................................... 111

6.3 Results ...................................................................................................................... 116

6.4 Discussion................................................................................................................. 121

6.5 Conclusions .............................................................................................................. 126

CHAPTER 7: TARGETED EDTA CHELATION THERAPY TO REVERSE

CALCIFICATION ........................................................................................................ 133

7.1 Introduction ............................................................................................................. 133

7.2 Materials and Methods ........................................................................................... 134

7.3 Results ...................................................................................................................... 147

7.4 Discussion................................................................................................................. 159

7.5 Conclusions .............................................................................................................. 164

CHAPTER 8: CONCLUSIONS AND RECOMMENDATIONS ............................. 174

Page 13: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XII

8.1 Conclusions .............................................................................................................. 174

8.2 Limitations of this project and recommendation for future work ..................... 176

CHAPTER 9: REFERENCES ..................................................................................... 179

SUPPLEMENTAL FILES ........................................................................................... 196

Page 14: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XIII

LIST OF TABLES

Table Page

Table 2.1. Types and characteristics of vascular calcification ......................................... 20

Table 2.2. Animal models of vascular calcification. ........................................................ 24

Table 2.3.Current or prospective therapies for treating vascular calcification. ................ 29

Table 2.4. Types and applications of chelation therapy. ................................................... 32

Table 2.5. EDTA’s binding constants to metal ions ........................................................ 33

Table 2.6. In vitro and animal studies for chelation therapy ............................................ 36

Table 2.7. Case studies and clinical trials of EDTA chelation therapy. ........................... 40

Table 2.8. Side effects of EDTA chelation therapy. ......................................................... 43

Table 2.9. Current opinions of EDTA chelation therapy for cardiovascular disease. ...... 45

Table 4.1. PCR primers used in the RT-PCR study. ......................................................... 62

Table 7.1. Characterization of nanoparticles. ................................................................. 148

Table 7.2. Organ distribution of fluorescent nanoparticles............................................. 154

Page 15: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XIV

LIST OF FIGURES

Figure Page

Figure 2.1. Structure of blood vessel. ................................................................................. 8

Figure 2.2. Structure of aortic elastin. ................................................................................. 9

Figure 2.3.Cut section of an artery showing atherosclerosis . .......................................... 11

Figure 2.4. Normal aorta and aorta with large abdominal aneurysm ............................... 13

Figure 2.5. H&E of a patient with MAC. ......................................................................... 15

Figure 2.6. Schematic illustrating four, non–mutually exclusive theories for vascular

calcification. ...................................................................................................................... 17

Figure 2.7. Osteo/chondrocytic differentiation of VSMC and vascular calcification ...... 18

Figure 2.8. Representative images for types of vascular calcification.. ........................... 21

Figure 2.9. Intra-luminal delivery devices ........................................................................ 47

Figure 2.10. Peri-adventital delivery methods .................................................................. 48

Figure 2.11. Novel sustained release vehicles .................................................................. 49

Figure 4.1. Characterization for calcified elastin .............................................................. 67

Figure 4.2. Expression of SMA and MHC in RASMCs cultured on hydroxyapatite and

calcified elastin at day 1 and day 7 ................................................................................... 70

Figure 4.3. Expression of osteogenic/chondrogenic markers in RASMCs cultured on

hydroxyapatite and calcified elastin elastin at day 1 and day 7. ....................................... 73

Page 16: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XV

List of Figures (Continued)

Figure Page

Figure 4.4. Expression of SMA and MHC in RASMCs after removal of calcified matrix.

........................................................................................................................................... 76

Figure 4.5. Expression of osteogenic/chondrogenic markers in RASMCs after removal of

calcified matrix ................................................................................................................. 77

Figure 4.6. Model of putative links between medial elastin-specific calcification and

chondro/osteoblast-like differentiation of VSMCs in the artery....................................... 82

Figure 4.7. Experimental design. ...................................................................................... 84

Figure 4.8. Graphical Abstract. ......................................................................................... 85

Figure 5.1. Demineralization of Hydroxyapatite. ............................................................. 94

Figure 5.2. Demineralization of calcified elastin .............................................................. 97

Figure 5.3. Demineralization of calcified human aorta. ................................................... 99

Figure 5.4. EDTA release profile from PLGA nanoparticles. ........................................ 101

Figure 5.5. Reversal of arterial calcification in vivo by EDTA ...................................... 102

Figure 5.6. Types of chlating agents and chemical structure. ......................................... 109

Figure 5.7. Surgery protocol and photos. ........................................................................ 109

Figure 6.1. Experimental protocol for CaCl2 injury model and EDTA treatment of rats..

......................................................................................................................................... 112

Page 17: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XVI

List of Figures (Continued)

Figure Page

Figure 6.2. Evaluation of aortic calcifications by stereomicroscope, histology and AAS.

......................................................................................................................................... 118

Figure 6.3. Ca quantification in serum and urine by AAS.............................................. 119

Figure 6.4. Bone integrity of EDTA-treated (study group) and untreated rats. .............. 121

Figure 6.5. H&E staining for rats’ kidneys. .................................................................... 127

Figure 6.6. One representative aorta’s Micro CT images from two groups. .................. 128

Figure 6.7. Three points bending test: before and after fracture. .................................... 129

Figure 6.8. Load – Displacement curve for three points bending. .................................. 130

Figure 6.9. Micro-CT for rats’ femoral bone. ................................................................. 131

Figure 6.10. Micro-CT for three slices of rats’ femoral bone. ........................................ 132

Figure 7.1. A: Schematic representation of antibody surface coating strategy for albumin

NPs. ................................................................................................................................. 141

Figure 7.2. Characterization for albumin NPs – optimization and function. .................. 150

Figure 7.3. Antibody binding and targeting efficacy in vitro ......................................... 151

Figure 7.4. Efficacy of EL-NPs targeting to damaged aorta in vivo. .............................. 153

Figure 7.5. Evaluation of aortic calcification for both systemic EDTA chelation therapy

and targeted EDTA chelation therapy. ............................................................................ 157

Page 18: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

XVII

List of Figures (Continued)

Figure Page

Figure 7.6. Possible side effects with systemic EDTA chelation therapy and targeted

EDTA chelation therapy. ................................................................................................ 159

Figure 7.7. H&E staining for rats’ kidneys (All five groups). ........................................ 166

Figure 7.8. Flow chart for preparation of EDTA loaded albumin NPs. .......................... 167

Figure 7.9. Graphical abstract to show nanoparticle structure and targeting and chelation.

......................................................................................................................................... 168

Figure 7.10. DiR standard curve and release profile in vitro. ......................................... 169

Figure 7.11. NPs bio-distribution present by organs. ..................................................... 171

Figure 7.12. NPs bio-distribution present by individual rats .......................................... 172

Figure 7.13. One representative aorta’s Micro CT images from all five groups. ........... 173

Page 19: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

1

CHAPTERS

CHAPTER 1: INTRODUCTION

Cardiovascular disease (CVD) refers to several types of conditions that affect the

heart and blood vessels. CVD is the leading cause of death in the world, responsible for

30% of all deaths globally, and 1 in 3 (over 800,000) deaths in the US [1]. Pathological

vascular calcification has been observed in many CVD and is recognized as a major risk

factor for various cardiovascular events including heart failure, pulmonary hypertension

(PTH), and death in chronic kidney disease (CKD) patients. Based on the location,

vascular calcification can be classified atherosclerotic intimal calcification (AIC), medial

arterial calcification (MAC) or Mönckeberg’s medial calcific sclerosis, aortic valve

calcification (AVC), and calcific uremic arteriolopathy(CUA) or calciphylaxis [2]. AIC

occurs in the context of atherosclerosis, associated with lipids, macrophages, and vascular

smooth muscle cells; whereas, MAC can exist independently of atherosclerosis and is

associated with elastic lamellae and vascular smooth muscle cells [3]. The work

presented here focuses on the mechanisms and reversal of MAC.

Currently the only option to treat vascular calcifications are surgical methods like

directional atherectomy to remove mineral physically and placement of stent grafts to

keep the artery open [4, 5]. These surgical methods are quite invasive and detrimental,

and there is no available alternative pharmacological treatment for reversal of vascular

calcification [6, 7]. One possible approach is EDTA chelation therapy. EDTA chelation

therapy most often involves the injection of disodium ethylene diamine tetraacetic acid

Page 20: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

2

(EDTA), a chemical that binds, or chelate ionic calcium, trace elements and other

divalent cations so as to reverse calcification. There are, however, contradictory opinions

for its success in improving cardiovascular health in patients.

Overall goal of our research is to understand mechanisms of elastin specific

MAC, to evaluate efficacy of chelating agents to reverse elastin specific MAC and their

possible side effects, and to develop a targeted delivery system to reverse elastin specific

MAC that can circumvent side effects.

Over the last decade it has been well established that vascular calcification is not a

passive deposition of mineral but an active process that resembles physiologic skeletal

mineralization in that it shows the presence of many bone proteins and

osteo/chondrogenic cells. It has been reported that the vascular calcification process is

mineral-ion dependent and can be caused by either osteogenic transdifferentiation of

VSMCs or reduced levels of mineralization inhibitors. However, it is unclear if

calcification occurs first and that triggers osteoblast-like differentiation of vascular

smooth muscle cells (VSMCs) or vice versa. It is still unclear if there exists a

chronological occurrence between the osteoblast-like differentiation of VSMCs and

passive deposition of hydroxyapatite on elastin, or if both processes occur simultaneously

causing the eventual calcification of arteries. We, for the first time, showed that in

response to the calcified matrix, for both hydroxyapatite crystals and calcified aortic

elastin, VMCs lost their smooth muscle lineage markers and underwent

chondroblast/osteoblast-like differentiation. Interestingly, this phenotypic transition was

Page 21: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

3

reversible and VSMCs restored their original linage upon reversal of calcification. Our

data supports the hypothesis that mineral deposition occurs first before osteogenic

transformation of VSMCs

EDTA chelation therapy has been touted as an effective treatment to reverse

vascular calcification; however, it has been controversial with opposing views [8-10]. At

present no systemic scientific studies prove that it could reverse cardiovascular

calcification or improve cardiovascular function [9]. Moreover, most chelating studies are

performed for atherosclerotic intimal calcification and no studies looked at whether such

therapy would work to reverse medical arterial calcification (MAC). First, we showed

that common chelating agents, such as disodium ethylene diamine tetraacetic acid

(EDTA), diethylene triamine pentaacetic acid (DTPA) and sodium thiosulfate (STS) can

remove calcium from hydroxyapatite (HA) and calcified tissues without damaging the

tissue architecture in vitro. EDTA and DTPA were more effective than STS. This paved a

way to test if such chelation can be used to reverse MAC in vivo.

Most of the EDTA chelation therapies include EDTA subcutaneous infusion, or

systemic application of EDTA solution at very high dose (around 40 ~ 50 mg/kg) [11-

17]. Many side effects have been found with such a high systemic dose of EDTA

chelation therapy including: renal toxicity [18-23] , hypocalcemia [14, 24, 25], and bone

loss [14, 26]. But results are quite controversial for side effects. On the basis of these

reports for EDTA chelation therapy and our own in vitro results showing its effectiveness

in reversing elastin-specific MAC, we performed an animal study to investigate whether

Page 22: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

4

or not systemic EDTA chelation therapy could reverse local medial aortic calcification

(MAC). A CaCl2 mediated abdominal aortic injury model was used to induce local aortic

calcification in abdominal aortic region in rats [27, 28]. We demonstrated that systemic

EDTA chelation therapy did not reverse local aortic calcification. Not only it did not

reverse MAC, but it also caused adverse events for serum and urine calcium, specifically

hypocalcemia and hypercalciuria. Thus, our data showed that systemic infusion of EDTA

may not be a right option for reversal of MAC.

In contrast to systemic EDTA chelation therapy, nanoparticle-based targeted

EDTA delivery to the calcified aorta could lower dosage required due to improved

bioavailability and sustained release at the site [29]. Thus, we tested if we can load

EDTA in albumin based nanoparticles and target them to the aortic calcification sites in

vivo. We show that the elastin antibody coated nanoparticles can be targeted to vascular

calcification sites [30]. Our in vitro and in vivo studies demonstrated that elastin

antibody coated and EDTA loaded albumin NPs reversed elastin specific MAC and did

not cause side effects such as hypocalcemia and bone loss typically seen in systemic

EDTA chelation therapy.

Organization of dissertation

1) In Chapter 2, we present comprehensive overview of:

Vascular architecture: normal anatomy of blood vessel and structure of aortic

elastin.

Page 23: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

5

Vascular disease: atherosclerosis, aortic aneurysms and medial arterial

calcification (MAC).

Vascular calcification: mechanisms. Types and characteristics, animal

models for investigating vascular calcification, current or prospective

therapies for treating vascular calcification.

Chelation therapy: types and applications of chelation therapy, in vitro and

animal studies of chelation therapy.

EDTA chelation therapy: case studies and clinical trials, side effects with

traditional systemic EDTA chelation therapy, current opinions of EDTA

chelation therapy for cardiovascular disease and targeted EDTA chelation

therapy.

Targeted EDTA chelation therapy: local drug delivery in the vasculature,

albumin nanoparticles as drug carriers for different application and targeted

EDTA chelation therapy using EDTA loaded albumin nanoparticles

2) Chapter 3 describes the project goals, scope, and rationale.

3) In Chapter 4, we investigate mechanisms of elastin specific MAC, specifically

cause-and-effect relationship between the elastin-specific MAC and osteoblast-

like differentiation of vascular smooth muscle cells (VSMCs).

4) In Chapter 5, we evaluate efficacy of chelating agents to reverse elastin specific

MAC. We show that chelating agents, such as disodium ethylene diamine

tetraacetic acid (EDTA), diethylene triamine pentaacetic acid (DTPA) and sodium

Page 24: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

6

thiosulfate (STS) can remove calcium from hydroxyapatite (HA) and calcified

tissues without damaging the tissue architecture.

5) In Chapter 6, we investigate systemic EDTA chelation therapy’s therapeutic

effects in reversal of MAC and possible side effects in rat model of MAC.

6) Finally in Chapter 7, we developed a targeted delivery system to reverse elastin

specific MAC and circumvent side effects associated with systemic EDTA

chelation therapy. This study proved the concept that elastin antibody coated and

EDTA loaded albumin NPs might be a promising nanoparticle protein

engineering therapy to reverse elastin-specific medial calcification.

Page 25: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

7

CHAPTER 2: LITERATURE REVIEW

2.1 Vascular architecture

2.1.1 Normal anatomy of blood vessel

The cardiovascular system consists of the heart, blood vessels, and the

approximately five liters of blood for humans. The heart is a muscular pumping organ

that circulates blood to the organs. There are two primary circuits in the human body: the

pulmonary circuit and the systemic Circuit. Pulmonary circuit consists of vessels that

carry blood from the heart to the lungs and back to the heart. Systemic circuit composed

of vessels that lead from the heart to all body parts (except the lungs) and back to the

heart.

Blood vessels are the body’s highways that allow blood to flow quickly and

efficiently from the heart to every region of the body and back again. The walls of

arteries and veins have three layers or tunics (Figure 2.1): 1) an outer layer (tunica

adventitia) of connective tissue with collagen fibers and fibroblast cells; 2) a middle layer

(tunica media), is mainly composed of smooth muscle cells and elastic fibers; 3) an inner

layer (tunica intima), is mainly composed of a monolayer of endothelial cells and a

basement membrane.

Page 26: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

8

Figure 2.1. Structure of blood vessel [31].

2.1.2 Structure of aortic elastin

Elastin is the predominant structural protein of the arteries comprising of ~ 50%

of the arterial extracellular matrix (ECM) [32]. Elastin is primarily synthesized during

embryogenesis, rarely undergoes remodeling and has a very low turn-over rate.

Page 27: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

9

Figure 2.2. Structure of aortic elastin. A: aorta stained by Verhoeff Van Gieson staining

10x magnification [33]; B: Depiction of elastic recoil [34]; C: Schematic representation

of elastin and microfibrils [35]; D: Structural features of elastin showing the repetitive

alternating sequence (square brackets) of β-hydrophobic domains and α-crosslinking

areas [36]; E: TEM of cross section of elastin and fibrillin fibers deposited by vascular

cells in culture [37]; F: TEM of longitudinal section of elastin. Elastin is stained in black

and is surrounded by microfibrils [38].

Page 28: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

10

Structure of elastin with different scale is shown in Figure 2.2. Verhoeff Van

Gieson histological staining shows elastin in aorta as black wavy structures in cross-

section. Elastic lamina in the artery is structured as sheets similar to lasagna. (Figure

2.2A). Elastin is endowed with high resilience capable to undergoing cyclic stretching

(Figure 2.2B). Elastic fibers are composed of 90% elastin core and 10 % microfibrils

(Figure 2.2C). Elastin is a biopolymer with tropoelastin as its single repeating

monomeric unit. The typical amino acid composition consists of non-polar amino acids

with few polar side chain residues. Elastin contains alternating hydrophobic segments and

alanine and lysine rich segments (Figure 2.2D) [36]. Under transmission electron

microscope, elastin appears either in cross-section as amorphous clumps exhibiting low

electron density measuring 100-800 nm at the core (Figure 2.2E) [37] or as

longitudinally-aligned, laterally-associating bundles of elastin (Figure 2.2F) [38].

2.2 Vascular diseases related to calcification

2.2.1 Atherosclerosis

Atherosclerosis is a disease of the arteries in which cholesterol deposited in the

wall of an artery, resulting in inflammation and narrowing of the arterial lumen channel

that may eventually impair blood flow. Atherosclerosis is characterized as combination of

tear in artery wall, cholesterol deposits, macrophage cell, macrophage foam cells, fat

deposits and hydroxyapatite crystals [39] (Figure 2.3).

Page 29: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

11

Figure 2.3. Cut section of an artery showing atherosclerosis [40].

The pathogenesis of atherosclerosis remains incompletely understood.

Accumulation of oxidized lipoproteins (oxLDL) within the vascular wall drives a related

immune response very early during the disease course. Such an immune response is self-

amplified and eventually escapes from physiologic control mechanisms [41].

Inflammation plays a large role in atherosclerosis. The process of inflammation regulates

the production of many chemokines and acute phase proteins. One of those acute phase

proteins is C-reactive proteins (CRP) [42]. CRP is a phylogenetically highly conserved

plasma protein, with homologs in vertebrates and many invertebrates, that participates in

the systemic response to inflammation. Recently, an association between minor CRP

elevation and future major cardiovascular events has been recognized [42].

Since atherosclerosis is an inflammatory disease, most treatments include

strategies to reduce inflammatory changes include interfere with the production of

reactive oxygen species or, more desirably, with protein kinase C and nuclear factor

kappa B (NF-kB) pathways [43]. From aspects of lifestyle and diet, a balanced diet

Page 30: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

12

should be low in saturated fatty acids and enriched in polyunsaturated fatty acids –n-3

fatty acids in particular [44]. And more uptake of antioxidants (such as vitamins E and C)

[45], and polyphenols present in green tea and in red wine [46] would also be helpful.

Some available pharmaceutics include: 1) anti-inflammatory drugs such as

glucocorticoids and non-steroidal anti-inflammatory drugs [47], and aspirin [48]; 2) MG-

CoA reductase inhibitors (statins). Statins competitively inhibit HMG-CoA reductase, the

rate-limiting enzyme in cholesterol synthesis. Statins also have non-lipid-decreasing

effects include partial agonism of PPARα and PPARγ [49]. PPARs are nuclear receptors

that, after ligand-induced activation, form a heterodimer with a ligand-activated retinoic

acid receptor.

Surgical methods currently used include: bypass graft surgery [50], stented or

non-stented balloon angiography [51]. However, neither of these methods reverses

calcification.

2.2.2 Aortic aneurysms

Aortic aneurysm is a local dilatation of the aorta with respect to the original

arterial size or the adjacent area of the artery (Figure 2.4). As a convention, arterial

diameter greater than 1.5 times the normal size or greater than 3 cm in diameter, is

considered aneurysmal, although the definition may vary somewhat by age and body

surface area [52].

The gross characterization of aortic aneurysms include locally dilated aorta,

atheromatous plaque deposition and mural thrombosis, whereas typical histological

Page 31: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

13

characterization include trans-mural inflammation, increased pro-inflammatory and

immune markers, extensive local proteolytic activity, apoptosis, local oxidative-stresses

and neovascularization.

Figure 2.4. Normal aorta and aorta with large abdominal aneurysm [53].

Currently advanced imaging techniques are used to detect and monitor aneurysms

include: ultrasonography, computed tomography (CT), and magnetic resonance imaging

(MRI). Ultrasonography is currently the safest, easiest and most economic examination

for the diagnosis of aneurysms. CT accurately demonstrates the size and extent of

aneurysms and also detects thrombus (crescent sign) effectively. MRI demonstrates

features without the need for a contrasting agent. MRI is completely non-invasive.

Currently there are two different methods to treat aneurysms in patients- an open

repair or an endovascular approach. There are no well-defined pharmacological therapies

for the treatment of aneurysms. Some available therapies have been tested including:

Page 32: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

14

statins [54], Beta androgenic blockers [55], tetracyclines [56], anti-platelet therapy [57],

anti-oxidant therapy- Vitamin E [58], and anti-inflammatory agents [59].

2.3 Vascular calcification

Cardiovascular disease (CVD) refers to several types of conditions that affect the

heart and blood vessels. CVD is the leading cause of death in the world, responsible for

30% of all deaths globally, and 1 in 3 (over 800,000) deaths in the US[1]. Pathological

vascular calcification has been observed in many CVD and is recognized as a major risk

factor for various cardiovascular events including heart failure, pulmonary hypertension,

and increased blood pressure [60]. Based on the location, vascular calcification can be

classified as intimal calcification, medial calcification, valvular calcification, and

calciphylaxis [2]. Many animal models have been developed to study mechanisms of

vascular calcification, however; there is only little literature available about preventing or

reversing vascular calcification. We are mostly concerned here with medial arterial

calcification (MAC) in the present work.

2.3.1 Medial arterial calcification (MAC)

Calcification of the media or medial artery calcification (MAC), also known as

Monckeberg’s sclerosis is a pathological ossification of the medial part of arteries

(Figure 2.5). Intimal calcification occurs in the context of atherosclerosis, associated with

lipid, macrophages and vascular smooth muscle cells, whereas MAC can exist

Page 33: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

15

independently of atherosclerosis and is associated with elastin and vascular smooth

muscle cells [3].

Figure 2.5. H&E of a patient with MAC. The arrow depicts calcium deposits within the

media of a peripheral artery [61].

MAC is frequently observed in patients with diabetes and is associated with

increased risk of nephropathy, retinopathy, lower limb amputation, and coronary artery

disease and all-cause mortality [62].

2.3.2 Mechanisms of vascular calcification

Pathologic calcification of cardiovascular structures, or vascular calcification, is

associated with a number of diseases including end-stage renal disease (ESRD) and

cardiovascular disease (CVD) [63].

Five different mechanisms for initiating vascular calcification are shown in

Figure 2.6 [63]. First, lack of or loss of inhibitors of mineralization, such as matrix Gla

protein (MGP), pyrophsophate in blood vessels normally expressed, have been shown

Page 34: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

16

cause spontaneous vascular calcification [64, 65]. Second, osteogenic mechanisms may

also play a role in vascular calcification. Many bone proteins such as osteopontin [66],

osteocalcin [67], and BMP-2 [68] have been identified in calcified vascular lesions.

Third, the link between osteoporosis and vascular calcification is identified and loss of

bone in osteoporosis has been correlated with higher calcium content in the arteries

(REF). Bone turnover causing circulating nucleational complexes has been proposed to

explain the link between vascular calcification and osteoporosis in postmenopausal

women [69]. Fourth, cell death produce phospholipid rich membranous debris and

apoptotic bodies that may serve to nucleate apatite which induce vascular calcification

[70]. Finally, via thermodynamic mechanisms (also referred to as “passive” mechanisms),

elevated Ca and P, promote apatite nucleation and crystal growth and would also

exacerbate vascular calcification initiated by any of the other mechanisms described

above. These mechanisms are not mutually exclusive and some or all can occur

simultaneously in vascular calcification.

Page 35: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

17

Figure 2.6. Schematic illustrating four, non–mutually exclusive theories for vascular

calcification [63]. 1) Loss of inhibition like matrix Gla protein (MGP), osteopontin

(OPN), fetuin, pyrophsophate, and others; 2) Induction of bone formation due to high

level of Pi, lipids, inflammatory cytokines, and others; 3) Circulating nucleational

complexes; 4) Cell death.

Current understanding of cause-and-effect relationship between

osteo/chondrocytic differentiation of VSMCs and vascular calcification is shown in

Figure 2.7 [71]. Normally, mesenchymal stem cells differentiate to adipocytes,

osteoblasts, chondrocytes, and vascular smooth muscle cells (VSMCs). In the setting of

chronic kidney disease, diabetes, aging, inflammation, and multiple other toxins, these

VSMC can dedifferentiate or transform into osteo/chondrocytic-like cells by upregulation

of transcription factors such as runt-related transcription factor 2 (RUNX-2) RUNX-2

and muscle segment homeobox 2 (MSX2). These cells lay down collagen and

noncollagenous proteins in the intima or media and incorporate calcium and phosphorus

Page 36: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

18

into matrix vesicles to initiate mineralization and further mineralize into hydroxyapatite.

What triggers this dedifferentiation of VSMCs is currently unknown.

Our research is testing the hypothesis that vascular calcification might precede

osteo/chondrocytic differentiation of VSMCs. Detailed experimental rationale is

discussed in Chapter 3.

Figure 2.7. Osteo/chondrocytic differentiation of VSMC and vascular calcification [71].

Abbreviations: CKD-Chronic kidney disease; PPi-pyrophosphate; MGP-matrix Gla

protein; OP-osteopontin.

2.3.3 Types and characteristics of vascular calcification

Based on histoanatomy or location, vascular calcification can be mainly classified

as four types (Table 2.1, Figure 2.8): atherosclerotic intimal calcification (AIC), medial

Page 37: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

19

arterial calcification (MAC) or Mönckeberg’s medial calcific sclerosis, Aortic valve

calcification (AVC), and calcific uremic arteriolopathy (CUA) or calciphylaxis.

AIC forms at the intimal layer of the blood vessel, and is mostly associated with

conditions of atherosclerosis, hyperlipidemia, and hypertension. ACI is characterized by

eccentric lumen-deformation, vessel remodeling, elastinolysis and increased oxidized

low-density lipoprotein (LDL) [68, 72-74].

MAC forms at the tunica media. MAC is mostly associated with type 2 diabetes

mellitus, end-stage renal disease (ESRD), hyperphosphatemia and Lower-extremity

amputations. MAC has circumferential calcium deposition in the arterial tunica media

along with the elastic lamina, adventitial inflammation, fibro-fatty expansion [75-79].

AVC forms at the aortic side of the leaflets and is usually associated with

hyperlipidemia, congenital bicuspid valve and rheumatic heart disease. AIC has fibro-

fatty expansion and inflammation of the lamina fibrosa, displaced and/or split elastic

lamina and nodular amorphous calcium phosphate accumulation via epitaxial deposition

[12, 80-82].

CUA refers to calcification formed in micro-vessels and is mainly associated with

end-stage renal disease (ESRD), and warfarin treatment. CUA is exhibited in arteriolar

(~ 0.6 mm diameter) medial calcification in dermal, pulmonary and mesenteric vessels.

CUA has fibroproliferative occlusion, secondary skin and dermal fat necrosis,

periarteriolar inflammation and bone morphogenetic protein 4 (BMP4) expression and

reduced Gla-modified matrix Gla protein (MGP) expression [83-86].

Page 38: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

20

Table 2.1. Types and characteristics of vascular calcification (Modified from

Demer(2008)[87], Towler(2008)[88])

Type Location Associated

conditions

Histopathology

and unique serum

biochemistry

References

Atherosclerotic

intimal

calcification(AIC)

Intimal Atherosclerosis;

Hyperlipidemia;

Osteoporosis;

Hypertension

Eccentric lumen-

deformation;

Vessel remodeling,

elastinolysis;

Increased oxidized

LDL

Boström(1993)[68]

Parhami(1997)[72]

Tintut(2000)[73]

Abedin(2004)[74]

Medial arterial

calcification(MAC)

a.k.a “Mönckeberg's

medial calcific

sclerosis”

Tunica

media

Type 2 diabetes

mellitus;

ESRD;

Hyperphosphatemia;

Amputation

Circumferential

calcium deposition

in the arterial tunica

media;

Adventitial

inflammation,

fibro-fatty

expansion;

Elevated circulation

OPG;

Matrix vesicle

mediated

calcification

Nelson(1988)[75]

Lehto(1998)[76]

Top(2002)[77]

Bas(2006)[78]

Al-Aly(2007)[79]

Aortic valve

calcification(AVC)

Aortic

face of

the

leaflets

Hyperlipidemia;

Congenital bicuspid

valve;

Rheumatic heart

disease

Fibro-fatty

expansion and

inflammation of the

lamina fibrosa;

Displaced and/or

split elastic lamina;

Rajamannan(2003)[80]

Katz(2006)[81]

O’Brien(2006)[12]

Rajamannan(2007)[82]

Calcific uremic

arteriolopathy(CUA)

a.k.a “calciphylaxis”

Micro-

vessels

ESRD;

Warfarin

Arteriolar(~ 0.6

mm

diameter)medial

calcification in

dermal, pulmonary

and mesenteric

vessels;

Fibroproliferative

occlusion,

secondary skin and

dermal fat necrosis;

Coates(1998)[83]

Janigan(2000)[84]

Wilmer(2002)[85]

Griethe(2002)[86]

BMP4: bone morphogenetic protein 4; MGP: matrix Gla protein; ESRD: end-stage renal disease; LDL:

low-density lipoprotein;

OPG: osteoprotegerin

Page 39: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

21

Figure 2.8. Representative images for types of vascular calcification. Top: photos or

radiograph. Scale bar: 1 cm. bottom: H&E staining images. Magnification: 100X, Scale

bar: 100 µm. A: Healthy non-calcified aorta as control; B: Severe atherosclerotic intimal

calcification; C: Medial arterial calcification; Top:Pelvic and lower extremity radiograph

shows extensive calcification of the femoral arteries; Bottom: Microphotography of

arterial wall with calcified (violet colour) atherosclerotic plaque; D: Aortic valve

calcification; E: Calcific uremic arteriolopathy; Calciphylaxis on the abdomen of a

patient with end stage renal disease. Markings are in cm. Images information and

resources: A (49 years old male patients) and B (Severe stherosclerotic patients, 75 years

old male patients) were from cadavers provided by Dr. Michael E. Ward at Greenville

Hospital System (Greenville, SC).

C:http://en.wikipedia.org/wiki/Monckeberg's_arteriosclerosis; D and E bottom: From

pathologist - Lang Lei at The Second Affiliated Hospital to Nanchang University (Jiang

Xi, P. R. China); E top: http://en.wikipedia.org/wiki/Calciphylaxis.

2.3.4 Animal models for investigating vascular calcification

Many animal models are developed to study vascular calcification. Table 2.2

summarizes these animal models based on specific treatment protocols, species studied,

and location of calcification. Treatments studied include calcium chloride (CaCl2) injury

of abdominal aorta [27, 28, 30, 89-91], local perfusion of pancreatic elastase [92-94],

balloon angioplasty[95], systemic delivery of warfarin plus vitamin K [30, 96, 97],

Page 40: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

22

Vitamin D [78, 98-100], nicotine plus vitamin D [101-104], indoxyl sulphate [105], and

many types of gene knockout models [64, 106-108].

The calcium chloride (CaCl2) injury model has been used for rat, mice, and rabbit

[27, 28, 30, 89-91]. Local damage to the abdominal aorta (or other regions like carotid

artery or thoracic aorta) is caused by direct application of CaCl2 resulting in chemical

damage of the tissue. This type of localized medial calcification is pathologically similar

to clinical cases and minimal injury to the arterial wall. In this model, adult rat aorta is

exposed and inflammation is induced by a single, periarterial application of calcium

chloride. This acute insult to the artery induces an accelerated and extensive calcification

accompanied by intense inflammation and severe elastin degradation [109].

The warfarin plus vitamin K model for rat typically uses 20 mg/kg/day of

warfarin in drinking water and 15 mg/kg/day of Vitamin K by subcutaneous injection

every other day for a total period of three weeks [30, 96, 97]. This treatment will cause

systemic calcification in the aorta, major arteries and heart valves for rats. Warfarin

inhibits activation of MGP (an important inhibitor of calcification), and vitamin K

restores normal coagulation time to offset the anticoagulant side effects of warfarin.

The vitamin D model for rat typically uses daily intraperitoneal injections of

vitamin D (1µg/kg) or supra-physiological vitamin D doses in diet (7.5 mg/kg) [78, 98-

100]. This treatment also causes systemic calcification in the aorta and major arteries.

The vitamin D model was used to mimic vascular calcification disease related to

osteoporosis and hyperparathyroidism secondary to chronic kidney disease. Toxicity of

Page 41: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

23

vitamin D is associated with hyperabsorption of calcium and phosphorus [100]. Vitamin

D exerts a stimulatory effect on vascular calcification through direct inhibition of the

expression of parathyroid hormone–related peptide (PTHrP) in VSMCs as an endogenous

inhibitor of vascular calcification [110].

Page 42: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

24

Table 2.2. Animal models of vascular calcification.

Protocol Species Mechanisms Location Characteristics References

CaCl2; 0.15M ~ 0.5 M

Single peri-

adenticial delivery

Rat/mice /rabbit

Chemical damage Abdominal aorta;

Carotid artery;

Thoracic aorta

Localized medial calcification similar to

clinical cases;

Local calcification at injury area

Basalyga(2004)[27] Ikonomidis(2003)[89]

Gertz(1988)[90]

Lai(2013)[91] Lei(2013)[28]

Sinha(2014)[30]

Warfarin; 20 mg/kg/day in

drinking water

Vitamin K; 15 mg/kg/day,

subcutaneous

injection every other day, 3 weeks

Rat Warfarin inhibit activation of MGP;

Vitamin K restore

normal coagulation time

Systemically in aorta, major

arteries and

heart valves

Systemically induce all arteries’ calcification;

Warfarin in diet or in

drinking water is safer than injection

Price(1998)[96] Bouvet(2007)[97]

Sinha(2014)[30]

Vitamin D;

daily

intraperitoneal injections, 1µg/kg;

Or supra-

physiological vitamin D doses

(7.5 mg/kg) in diet

Rat Hyperabsorption of

calcium and

phosphorus

Systemically in

aorta and major

arteries

Currently used to mimic

osteoporosis and

hyperparathyroidism secondary to chronic

kidney disease

Boström(2000)[98]

Price(2001) [99]

Bas(2006)[78] Zittermann(2008)[100]

Nicotine; 25 mg/kg, 5

ml/kg,

oral

administration;

Mostly used

combined with vitamin D

Rat Nicotine amplifies the calcifying effects

of Vitamin D

Systemically in aorta and major

arteries

Associated with advancing age;

Accompanied with

systolic hypertension and

LV concentric

hypertrophy

Niederhoffer(1997)[101] Pan(2004)[102]

Park(2008)[103]

Zhou(2009)[104]

Indoxyl sulphate;

200 mg/kg in water

Rat Uremic toxins Aorta

Increase thickness of the

arcuate aorta, thoracic aorta and abdominal aortas

Adijiang(2008)[105]

Elastaste;

Intraluminal delivery,

5.23 U/mg

Swine/

rabbit

Chemical damage Local

abdominal aorta;

Associated with medial

calcification; Reproducibility is

controversial

Marinov(1997)[92]

Carsten(2001)[93] Ding(2006)[94]

Balloon angioplasty

Rabbit Mechanical damage Local aorta Mainly used to study restenosis

Gadeau(2001)[95]

Gene knockout

model;

FGF23 deficient;

Klotho deficient;

MGP knockout; OPG knockout;

Fetuin A knockout

Mice Gene knockout to

specific gene related

to regulation of

vascular calcification

Systemically in

arteries

Showed specific role of

proteins in the

calcification process

Kuro-o(1997)[106]

Luo(1997)[64]

Schäfer(2003)[107]

Razzaque(2006)[108]

FGF23, Fibroblast growth factor 23; LV: Left ventricular; MGP, Matrix Gla protein; OPG, Osteoprotegerin;

Page 43: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

25

The nicotine plus vitamin D model for rat uses combination of vitamin D with

nicotine (25 mg/kg, 5 ml/kg) given by oral administration [101-104]. This treatment

causes systemic calcification in the aorta and major arteries. Nicotine is added in

conjunction to the vitamin D to amplify the calcification processes. The nicotine plus

vitamin D model mimics calcification disease associated with advancing age and is

accompanied by systolic hypertension and left ventricular (LV) concentric hypertrophy.

The indoxyl sulphate (IS) model for rat uses 200 mg/kg of indoxyl sulphate in

drinking water [105]. The IS model showed increase thickness of the aorta. IS promotes

aortic wall thickening and aortic calcification with colocalization of osteoblast - specific

proteins [105]. Administration of IS to 5/6 nephrectomized rats caused glomerular

sclerosis in the remnant kidney with a decline in renal function [111]. Furthermore, IS

stimulated transcription of genes related to renal fibrosis [112].

The elastase model has been studied in swine and rabbit [92-94]. By intraluminal

infusion of elastaste after surgical exposure of aorta, an important number of necrotic

lesions were observed, and they were associated with the development of calcium

deposits by 3 weeks [92]. Elastase degrades elastin in elastic lamina of arteries, which in

turn induces local calcification for abdominal aorta. Many surgical modified techniques

have been investigated to get reproducible elastase-induced aneurysms related

calcification, such as direct exposure of the origin of the right common carotid artery

(RCCA) with placement of a temporary aneurysm clip instead of an occlusion balloon for

Page 44: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

26

isolation of the artery, and more extensive dissection of the RCCA, down to its origin

from the brachiocephalic artery [94].

The balloon angioplasty model for rabbit uses mechanical damage to induce local

calcification of abdominal aorta [95]. The aortic injury was rapidly followed by calcified

deposits that appeared in the media as soon as 2 days after injury and then accumulated in

zipper-like structures. This rapid aortic calcification model is good to study mechanisms

involved in the initiation step of the aortic calcification process but mainly related to

restenosis [45].

Many types of gene knockout models for mice have been studied including

Fibroblast growth factor 23(FGF23) deficient, Klotho deficient, matrix GLA protein

(MGP) knockout, OPG (Osteoprotegerin) knockout and fetuin A knockout model[64,

106-108]. Knocking out specific genes related to regulation of vascular calcification will

cause systemic calcification in arteries and will show the specific role of proteins in the

calcification process. Kuro-o et al. in 1997 showed that defect in klotho gene expression

in the mouse results in a syndrome that resembles human ageing, including a short

lifespan, arteriosclerosis, skin atrophy etc. The klotho gene encodes a membrane protein

that shares sequence similarity with theb-glucosidase enzyme [106]. Luo et al. in 1997

showed that mice that lack MGP develop to term but die within two months as a result of

arterial calcification which leads to blood vessel rupture and concluded that MGP is the

first inhibitor of calcification of arteries and cartilage to be characterized in vivo [64].

Schäfer et al. in 2003 identified the serum protein α2–Heremans-Schmid glycoprotein

Page 45: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

27

(Ahsg, also known as fetuin-A) as an important inhibitor of ectopic calcification acting

on the systemic level. Ahsg-deficient mice were phenotypically normal, but develop

severe calcification of various organs on a mineral and vitamin D–rich diet [107].

Razzaque et al. in 2006 foung that FGF23 deficient mice exhibited numerous

biochemical and morphological features consistent with premature aging-like phenotypes,

along with severe atherosclerosis, widespread soft tissue calcifications [108].

2.3.5 Current or prospective therapies for treating vascular calcification

Current or prospective therapies for treating vascular calcification are summarized

in Table 2.3 (Modified from Kapustin (2009) [6], Wu (2013) [7]). These treatments are

often existing therapies for related conditions and could be classified as three categories

based on different targets and mechanisms of action: chronic kidney disease and

osteoporosis therapies, cardiovascular disease therapies, and experimental therapies.

Therapies for treating vascular calcification associated with chronic kidney

disease (CKD) and osteoporosis include: 1) treatments using bisphosphonates, sevelamer,

calcimimetics and thyroidectomy so as to target mineral imbalance, and 2) sodium

thiosulfate (STS) as a vasodilator, antioxidant, and calcium chelator [113-118]. The

treatments with bisphosphonates, sevelamer, calcimimetics and thyroidectomy were

proposed to maintain Ca and P serum levels, inhibit initialization and growth of calcium

apatite crystals and prevent osteo/chondrogenic transition of vascular smooth muscle cell

(VSMC). However, these treatments had complications and limitations such as crosstalk

with bone metabolism and require strict dialysis protocols; [116, 117, 119-122]. STS

Page 46: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

28

treatment could improve local circulation, reduce inflammation and calcification. STS is

an established treatment for calciphylaxis. However, STS also has possible side effects

such as hypocalcemia and bone loss [119].

Most common therapies for treating vascular calcification associated with

cardiovascular diseases are statins. Statins are a group of drugs that help to lower serum

cholesterol by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA)

reductase in the liver, to help prevent the formation of atherosclerotic lesions. Statins

have been used to lower serum cholesterol and reduce arterial calcification [123-125].

Statin treatments prevent inflammation and increase VSMC survival and viability.

Callister et al. in 1998 demonstrated that treatment with HMG-CoA reductase inhibitors,

or statins can reduce the calcium-volume score and volume of calcified plaque in the

coronary arteries [123]. A retrospective study of 174 patients with mild to moderate

calcific aortic stenosis was conducted by Novaro et al. in 2001 [124]. Results showed that

statin-treated patients had reduced aortic stenosis progression compared with those not

treated with a statin [124]. Although statins could prevent atherosclerosis, it is ineffective

for diabetic CKD [125]. Besides, statin treatments had side effects with the immune

system and tissue remodeling [126, 127]. Statins were found to suppress the induction of

Major histocompatibility complex class II (MHC-II) molecules, which affect the immune

response and organ rejection after transplantation [126].

Page 47: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

29

Table 2.3.Current or prospective therapies for treating vascular calcification(Modified

from Kapustin(2009)[6], Wu(2013)[7]).

Treatment Mechanisms of action Complication and limitation References

Chronic kidney disease and osteoporosis therapies

Bisphosphonates

Sevelamer Calcimimetics

Thyroidectomy

Target mineral imbalance;

Maintain Ca and P serum levels; Inhibition of initialization and

growth of the calcium apatite

crystal;

Side effects with bone metabolism;

Strict dialysis protocols

Hafner(1995)[113]

Asmus(2005)[114] Russell(2007)[115]

O'Neill(2008)[116]

O'Neill(2010)[117] Raggi(2011)[118]

Sodium thiosulfate A vasodilator, antioxidant, and

calcium chelator;

Improves local circulation,

reduces inflammation and calcification

Established treatment for

calciphylaxis;

Possible side effects such as

hypocalcemia and bone loss

Pasch(2008)[119]

O'Neill(2008)[116]

O'Neill(2010)[117]

Adirekkiat(2010)[120] Mathews(2011)[121]

Ong(2012)[122]

Vitamin K Increases activation of circulating MGP, an

inhibitor of calcification

Only worked for patients have vitamin K deficiency

Spronk(2004)[128] Holden(2010)[129]

Schlieper(2011)[130]

Westenfeld(2012)[131]

Cardiovascular disease therapies

Statins Prevent inflammation;

Increase VSMC survival and

viability

Side effects with immune system and

tissue remodeling;

Could prevent atherosclerosis but ineffective for diabetic CKD

Callister(1998)[123]

Novaro(2001)[124]

Wanner(2005)[125] Mach(2004)[126]

Greenwood(2006)[127]

Experimental therapies

Carbonic anhydrase II Induction of calcium deposit

resorption

Side effects with bone metabolism Rajachar(2009)[132]

Osteoclast-mediated demineralization

Main mineral-resorbing cells in the body;

Resorb abnormal minerals in

vascular calcification

Cells only migrated to the adventitial layer of the artery;

Successful delivery of the osteoclasts is

still a big challenge

Simpson(2007)[133]

EDTA Directly remove calcium from

calcified tissue

Binds serum calcium to reduce calcification;

Chelate toxic heavy metals;

Normalize calcium metabolism by reactivation of enzymes

inhibited by toxic heavy metals;

Anti-nanobacteria; Remove Fe and Cu to protect cell

membrane

Passible side effects including renal

toxicity, hypocalcemia, bone loss and

burning at injection site or along course

Bolick(1961)[134]

Maxwell(1963)[135]

Lufkin(1983)[136] Olszewer(1990)[137]

Guldager(1993)[14]

Cranton(2001)[138] Maniscalco(2004)[139]

Cavallotti(2004)[13]

Silay(2007)[140] Lamas(2013)[17]

Lei(2013)[28]

Treatments in italic type: clinical trials and are currently used; Treatments as listed not by italic type: hypothetical and experimental.

MGP: Matrix gla protein; VSMC: vascular smooth muscle cell

Page 48: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

30

Finally, many experimental therapies were tested in animal models including: 1)

Carbonic anhydrase II [132]; 2) Osteoclast-mediated demineralization [133]; and 3)

EDTA chelation therapy [135-137] [13, 14, 138-140] [17]. Carbonic anhydrase II had

shown an induction of calcium deposit resorption but it had side effects on bone

metabolism [132]. Osteoclasts are the only mineral-resorbing cells in the body and

osteoclasts delivery was tried as a method to resorb abnormal minerals in vascular

calcification. However, the Osteoclast-mediated demineralization method showed that

cells only migrated to the adventitial layer of the artery and successful delivery of the

osteoclasts still remains a challenge [133].

2.4 Chelation therapy

Chelation therapy most often involves the injection of EDTA, a chemical that

binds, or chelates ionic calcium, trace elements, and other divalent cations [141]. At

present, whether or not chelation therapy reverses vascular calcification is quite

controversial [9]. There are many proposed mechanisms for EDTA chelation therapy to

reverse calcification. EDTA can directly remove calcium from calcified tissue [28, 134].

EDTA can bind serum calcium to reduce calcium levels and that way can prevent mineral

augmentation. It can also chelate toxic heavy metals and normalize calcium metabolism

by reactivation of enzymes inhibited by toxic heavy metals [135-138]. EDTA also been

shown as an anti-nanobacteria and can remove Fe and Cu to protect the cell membrane

[13, 139, 140]. However, systemic side effects including renal toxicity [18-23] ,

hypocalcemia [14, 24, 25], bone loss [14, 26] prevent it from being an accepted form of

treatment for vascular calcification. If one can reduce systemic side effects, EDTA

Page 49: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

31

chelation therapy might still be a promising treatment for diseases of vascular

calcification. Site-specific targeted drug delivery has advantages such as lower toxicity,

higher therapeutic effect, thus one can envision such therapies for resorbing mineral from

arteries.

2.4.1 Types and applications of chelation therapy

Many types of chelators have been studied for medical applications including:

EDTA [17, 24, 142-144], calcium disodium EDTA (calcium disodium versante, CaNa2-

EDTA) [145-147], STS [120, 121, 148-151], diethylene triamine pentaacetic acid (DTPA)

[152, 153], dimercaprol (British anti-Lewisite; BAL) [145, 154], Dimercaptosuccinic

acid (DMSA) [145, 155], dimercapto-propane sulfonate (DMPS) [156, 157],

penicillamine [145, 158], deferoxamine and deferasirox [159, 160]. Current types and

applications of chelation therapy based on type of chelator are summarized in Table 2.4.

Page 50: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

32

Table 2.4. Types and applications of chelation therapy.

Type of chelator Medical applications References

EDTA Hypercalcemia;

Prevent calcification of fixed tissue;

Cardiovascular disease include heart disease

and atherosclerosis

Spencer(1956)[24]

Clarke(1956)[142]

Robert(1980)[143]

Sucu(2006)[144]

Lamas(2013)[17]

Calcium disodium EDTA

(calcium disodium versenate;

CaNa2-EDTA)

mercury and lead poisoning Trevor(2007)[145]

Saxena(2004)[146]

Leckie(1958)[147]

Sodium thiosulfate(STS) Antioxidant;

Cyanide toxicity;

Skin diseases like acne and pityriasis

versicolor;

Carboplatin and cisplatin-induced oto- and

nephrotoxicity;

Calcific uremic arteriolopathy(CUA) a.k.a

“calciphylaxis”;

Decrease vascular calcification in ESRD;

Delay progress of CAC in hemodialysis

patients;

Hayden(2005)[148]

Dickey(2005)[149]

Pouyatos(2007)[150]

Hayden(2008)[151]

Mathews(2011)[121]

Adirekkiat(2010)[120]

DTPA MRI contrasting agents;

Treatment of radioactive materials such as

plutonium, americium, and other actinides

Caravan(1999)[152]

Brown(2012)[153]

Dimercaprol

(British anti-Lewisite; BAL)

Lewisite poisoning (for which it was

developed as an antidote);

Arsenic, mercury, lead poisoning

Goldman(1989)[154]

Trevor(2007)[145]

Dimercaptosuccinic acid

(DMSA)

Arsenic, mercury, lead poisoning Archbold(2004)[155]

Trevor(2007)[145]

Dimercapto-propane sulfonate

(DMPS)

severe acute arsenic, mercury poisoning Aposhian(1990)[156]

Gonzalez-

Ramirez(1998)[157]

Penicillamine Mainly in copper toxicity of Wilson's

disease;

Occasionally adjunctive therapy in: gold

toxicity, arsenic poisoning, lead poisoning

rheumatoid arthritis

Peisach(1969)[158]

Trevor(2007)[145]

Deferoxamine and Deferasirox

Acute iron poisoning;

Iron overload

Moeschlin(1964)[159]

Westlin(1966)[160]

CAC: coronary artery calcification; DTPA: diethylene triamine pentaacetic acid; ESRD: End-stage renal

disease; MRI: Magnetic resonance imaging

Page 51: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

33

EDTA has been used in hypercalcemia, prevention of calcification for fixed tissue,

and as a treatment for cardiovascular diseases including atherosclerosis [17, 24, 142-144].

Tables 2.5 shows EDTA’s binding constants to metal ions (modified from Halstead

[103]), Rozema [104]). Higher binding constant indicates more potential of binding to the

metal ion. Clearly, EDTA binds strongly with iron, mercury and lead.

Table 2.5. EDTA’s binding constants to metal ions(modified from Halstead (1979[161]),

Rozema(1997)[162])

Metal cation Fe+++ Hg++ Cu++ Pb++ Ni++ Zn++ Cd++ Co++ Al+++ Fe++ Mn++ Ca++ Mg++

Log K 25.1 21.8 18.8 18.5 18.0 16.5 16.5 16.3 16.1 14.3 13.7 10.7 8.7

Calcium disodium EDTA (calcium disodium versenate, CaNa2-EDTA) [145-147]

has been used as treatment in mercury and lead poisoning and has been approved by US

Food and Drug Administration (FDA). STS has been used as an antioxidant, treatment

for cyanide toxicity, skin diseases like acne and pityriasis versicolor, carboplatin and

cisplatin-induced oto- and nephrotoxicity, calcific uremic arteriopathy(CUA) a.k.a

“calciphylaxis”. STS treatment has been shown to decrease vascular calcification in

ESRD and delay progress of CAC in hemodialysis patients [120, 121, 148-151].

DTPA has been used as magnetic resonance imaging (MRI) contrasting agents,

treatment of radioactive materials such as plutonium, americium, and other actinides [152,

153]. Dimercaprol (British anti-Lewisite; BAL) has been used for Lewisite poisoning (for

which it was developed as an antidote) and arsenic, mercury, lead poisoning [87, 96].

DMPS also has been used for severe acute arsenic, mercury poisoning [98, 99].

Page 52: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

34

Penicillamine is mainly used in copper toxicity of Wilson's disease and occasionally

adjunctive therapy in: gold toxicity, arsenic poisoning, and lead poisoning rheumatoid

arthritis [87, 100]. Deferoxamine and deferasirox is mainly used in acute iron poisoning

and iron overload [101, 102].

2.4.2 In vitro and animal studies of chelation therapy for vascular

calcification

Few studies have been reported for chelation therapy using EDTA [134] [11, 13,

21, 28, 144, 163-165] and STS [119, 166] to prevent or reverse calcification (Table 2.6).

It has been shown in vitro that EDTA could remove calcium from human

calcified tissue. A modified form of EDTA, NH4-EDTA, provided a quantitative method

of decalcifying tissue [134]. EDTA also has been used as a pre-treatment for

bioprosthetic materials such as collagen-elastin matrix (CEM) and bovine pericardium

specimens and significantly decreased the calcification of these tissues in a rat subdermal

model [163] [144, 164]. One study of the mechanism of renal vacuologenesis induced by

EDTA systemic infusion (jugular vein injection, 1000 mg/kg, disodium Ca-EDTA)

showed EDTA-induced vacuologenesis is a reflection of the induction of pinocytosis by

the chelate [21].

EDTA treatment by direct infusion (50 mg/kg, 15 mg/minutes, total 20 infusions)

showed significantly less calcium in the aorta of the EDTA infused animals in an

atherosclerotic rabbit model (cholesterol diet and Vitamin D3 injection) [11]. However,

O'Brien et al. shown that EDTA treatment (40 mg/kg, daily intravenous injection, 4

Page 53: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

35

weeks) caused a 74% increase in triglyceride levels and no benefit on arterial lesions in

an atherosclerotic, obese, diabetic rat model [165]. Cavallotti et al. showed that EDTA

treatment (40 mg/kg or 120 g/3 kg rabbit, daily intravenous injection, 10 days) showed a

marked decrease of the calcification in a rabbit calcification model [13]. We recently

compared chelating efficacy EDTA, DTPA and STS (1–10 mg/mL) to remove calcium

from hydroxyapatite (HA), calcified elastin and aortic tissue and showed that EDTA and

DTPA could effectively remove calcium from HA and calcified tissues, while STS was

not effective [28]. We further showed that EDTA-loaded PLGA nanoparticle pellets (30

mg) when placed periadventitially on calcified abdominal aorta could also reverse local

vascular calcification in vivo [28].

Page 54: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

36

Table 2.6. In vitro and animal studies for chelation therapy

Chelating

agent

Study design Results References

EDTA NH4-EDTA and EDTA were used to remove calcium from human calcified tissue

Provided a quantitative method of decalcifying tissue

Bolick(1961)[134]

SD rats, n=5 in each group; 1000 mg/kg of

disodium Ca-EDTA; jugular vein injection;

Renal toxicity Schwartz(1970)[21]

Rat subdermal model; Collagen-elastin matrix

(CEM) were implant after pretreatment with various conditions of ethanol and EDTA

Significantly decreased the

calcification rate of CEM

Singla(2003)[163]

Rat subdermal model; Pericardium specimens were

treated with PBS containing 100 µg/ml EDTA

EDTA was useful in the

attenuation of calcification

Sucu(2006)[144]

Rat subdermal model; Freshly excised bovine

pericardium fixed in PBS solution containing 11%

EDTA for a period of 48 h (30 ml/g tissue)

Calcium content of EDTA-

treated pericardium was

significantly lower

Sucu(2004)[164]

Atherosclerotic rabbit model by cholesterol diet and

Vitamin D3 injection;

EDTA: 50 mg/kg, 15 mg/minutes, total 20 infusions

Significantly less calcium in

the aortas of the EDTA

infused animals

Kaman(1990)[11]

Atherosclerotic, obese, diabetic rat model; EDTA: 40 mg/kg, daily intravenous injection, 4

weeks

No benefit on arterial lesions; But produced in a 74%

increase in triglyceride levels

O'Brien(2000)[165]

Rabbit calcification model by a single dose of 8 mg

of dihydro-tachysterol(synthetic vitamin D analog); EDTA: 40 mg/kg(120 g/3 kg rabbit), daily

intravenous injection, 10 days

Marked decrease of the

calcification

Cavallotti(2004)[13]

EDTA, DTPA and STS(1–10 mg/mL) were used to remove calcium from hydroxyapatite calcified

tissue;

EDTA-loaded PLGA nanoparticle pellets (30 mg) were placed periadventitially on calcified abdominal

aorta

EDTA and DTPA could effectively remove Ca from

HA and calcified tissues,

while STS was not effective

Lei(2013)[28]

Sodium thiosulfate(S

TS)

Uremic rat model by adenine-induced nephropathy; STS: 0.4 g/kg intraperitoneally 3 times a week;

Prevent vascular calcifications in

uremic rats;

Side effects such as bone loss and transiently lowered

ionized calcium in blood

plasma

Pasch(2008)[119]

Aortic calcification model and hydroxyapatite formation in vitro;

STS: 1-2 mM;

Aimed to determine effect of thiosulfate on vascular calcification and hydroxyapatite formation

Thiosulfate inhibited vascular calcification through a direct

extracellular effect related to

cellular injury

O’Neill(2012)[166]

One recent study by O’Neill et al. using STS aimed to determine the effect of

thiosulfate on vascular calcification and hydroxyapatite formation using an aortic

Page 55: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

37

calcification model and hydroxyapatite formation in vitro [166]. Their results showed that

thiosulfate prevented calcification of injured or devitalized aortas but not uninjured aortas.

Another animal study using systemic STS (0.4 g/kg intraperitoneally 3 times a week)

showed that STS treatment could prevent vascular calcifications in a uremic rat model by

adenine-induced nephropathy [119]. However, the STS treatment had side effects such as

bone loss and transiently lowered ionized calcium in blood plasma.

2.5 EDTA chelation therapy

2.5.1 Case studies and clinical trials of EDTA chelation therapy

Many case studies and clinical trials have been conducted for EDTA chelation

therapy (Summarized in Table 2.7). Results of all these studies are quite controversial.

On the positive side there are several studies that show improvement with chelation

therapy. Clarke et al. in 1956 [142] reported a case study of EDTA chelation therapy (5

g EDTA, intravenous infusion) with 20 patients with coronary heart disease showing 19

patients with improved symptoms post treatment with EDTA. They continued their

research with another bigger case study with 700 patients afflicted by coronary heart

disease and showed that 87% of the patients had improved symptoms post treatment with

EDTA [167]. Olszewer et al. in 1990 [137] did a pilot double blinded study with EDTA

chelation therapy (1.5 g EDTA, 20 intravenous infusion) with 10 patients (all male, aged

41 to 53 years) diagnosed with peripheral vascular disease. Results showed that EDTA

chelation therapy had substantial improvements for the walking test and master exercise

test. Rudolph et al. in 1991 [168] reported a case study with 30 patients diagnosed with

Page 56: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

38

atherosclerotic vascular disease. EDTA chelation therapy (3 g EDTA, 30 intravenous

infusions, and 10 months) was shown to decrease intra-arterial obstruction and reduce in

stenosis post treatment with EDTA. Hancke et al. in 1993 [169] reported a case study

with 470 patients and showed that after EDTA chelation therapy (50 mg/kg, 30

intravenous infusions, 3-4 months) more than 80% of the patients had objective evidence

for improvement post treatment with EDTA. Maniscalco et al. in 2004 [139] reported a

case study with 100 patients who had stable coronary artery disease (CAD) using ComET

therapy, which is composed of (1) nutraceutical powder, (2) tetracycline HCl, and (3)

EDTA 1.5 g taken in a rectal suppository base. After ComET therapy, coronary artery

calcium (CAC) scores decreased. Shoskes et al. in 2005 [170] reported a case study with

16 male patients with recalcitrant chronic prostatitis/chronic pelvic pain syndrome(CPPS)

showing significant improvement of symptoms in the majority of men after ComET

therapy. Cacchio et al. in 2009 [171] reported a case study with 80 patients with

radiographically verified calcific tendinitis of the shoulder and after single-needle

mesotherapy (1 ml of 15%disodium EDTA) calcifications were greatly decreased.

However, there were four published randomized trials of EDTA chelation therapy

therapy that are not completely consistent with the aforementioned case studies. The first

randomized trial was done by Guldager et al. in 1993 [14] with a total of 54 patients (15

women and 39 men, aged 41 to 81 years) with intermittent claudication. Patients

underwent EDTA chelation therapy (3 g EDTA, 20 intravenous infusion, 5-9 weeks).

However, results in this study showed no differences in groups in any clinically relevant

parameter and the EDTA chelation therapy was accompanied by bone loss. A study by

Page 57: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

39

Van et al. in 1994 [15] with 32 patients with intermittent claudication and also showed

that EDTA chelation therapy (3 g EDTA, intravenous infusion) had no significant

beneficial effects in patients with intermittent claudication but had some improvement in

resting ankle-brachial index. Knudtson et al. reported in 2002 [16] a case with 84 patients

who were at least 21 years old with coronary artery disease (CAD). This study showed

that EDTA chelation therapy (40 mg/kg EDTA, intravenous infusion) had no evidence to

support a beneficial effect. A recently study conducted by Lamas et al. in 2013 and

funded by NIH [17] with 1708 patients with myocardial infraction showed that EDTA

chelation therapy (50 mg/kg EDTA, intravenous infusion , one time a weeks, 40 weeks)

would modestly reduce the risk of adverse cardiovascular outcomes but the data did not

reach statistical significance. Moreover, this study did not highlight calcium score data.

Page 58: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

40

Table 2.7. Case studies and clinical trials of EDTA chelation therapy.

Reference Sample size Study design Results

Clarke(1956)[142] 20 patients; with coronary heart disease

5 g EDTA, intravenous infusion 19 improved, 1 died

Clarke(1960)[167] 700 patients;

with coronary heart disease

NA 87% improved

Olszewer(1990) [137] 10 patients(all male, aged 41

to 53 years); pilot double blinded study;

with peripheral vascular

disease;

1.5 g EDTA, 20 intravenous infusion

Substantial improvements for

walking test, master exercise test

Rudolph(1991) [168] 30 patients;

with atherosclerotic vascular

disease

3 g EDTA, 30 intravenous infusions,

10 months

Intra-arterial obstruction

decreased;

Reduction in stenosis

Guldager(1993) [14] Total 54 patients(15 women

and 39 men, aged 41 to 81

years); randomized controlled trial;

with intermittent

claudication

3 g EDTA, 20 intravenous infusion, 5-

9 weeks

Accompanied by bone loss

Hancke(1993) [169] 470 patients 50 mg/kg EDTA, 30 intravenous

infusions, 3-4 months

more than 80% had objective

evidence for improvement

Van(1994)[15] 32 patients; randomized controlled trial;

with intermittent claudication

3 g EDTA, intravenous infusion No significant beneficial effects in patients with intermittent

claudication;

Some improvement in resting ankle-brachial index

Knudtson(2002) [16] 84 patients(at least 21 years

old); randomized controlled trial;

with CAD

40 mg/kg EDTA, intravenous

infusion;

No evidence to support a

beneficial effect ; Positive treadmill test for ischemia

Maniscalco(2004)[139] 100 patients; with stable CAD and positive

CAC scores

ComET therapy was composed of (1) Nutraceutical Powder (2) Tetracycline

HCl;(3) EDTA 1.5 g taken in a rectal

suppository base

CAC scores decreased during ComET therapy trial

Shoskes(2005) [170] 16 male patients;

with recalcitrant chronic prostatitis/chronic pelvic pain

syndrome(CPPS)

ComET therapy Significant improvement in the

symptoms of recalcitrant CPPS in the majority of men

Cacchio(2009) [171] 80 patients;

with radiographically verified calcific tendinitis of

the shoulder

1 ml of 15% EDTA, single-needle

mesotherapy

Calcifications greatly decreased

Lamas(2013)[17] 1708 patients; Randomized controlled trial;

myocardial infraction

50 mg/kg EDTA, intravenous infusion , one time a weeks, 40 weeks

Modestly reduced the risk of adverse cardiovascular

outcomes; Did not highlight

calcium score data

CAD: coronary artery disease; CAC: coronary artery calcium; CPPS: chronic pelvic pain syndrome; NA: not available

Page 59: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

41

2.5.2 Side effects with traditional systemic EDTA chelation therapy

Many side effects have been found with traditional systemic EDTA chelation

therapy (Table 2.8), which include but are not limited to: renal toxicity [18-23] ,

hypocalcemia [14, 24, 25], bone loss [14, 26] and burning at injection site or along course

of vein [25].

The most important potential adverse event with EDTA chelation therapy is renal

toxicity [18-23]. Holland et al. in 1953 [18] reported that two instances of renal tubular

vacuolization following administration of sodium-EDTA. Schwartz et al. in 1966 [19]

showed that vacuolization also occurred and part of the chelate enters the cells via

pinocytosis post EDTA treatment. Doolan et al. in 1967 [20] also showed tubular

vacuolization produced in the rat by intraperitoneal injection of EDTA daily for 10 days;.

Schwartz et al. in 1970 [21] showed renal tubular cell vacuolization in the outer renal

cortex after EDTA chelation therapy (1000 mg/kg of disodium Ca-EDTA; Jugular vein

injection). Oliver et al. in 1984 [23] reported that patients have renal failure accompanied

with underlying renal disease after EDTA chelation therapy. However, McDonough et al.

in 1982 [22] did a study with 383 patients diagnosed with occlusive arterial disease and

showed that EDTA infusion (3 g EDTA; 10 intravenous infusion; 5 days between

infusions) is not nephrotoxic.

The second possible adverse event is hypocalcemia [14, 24, 25]. Spencer et al. in

1956 [24] showed transient hypocalcemia after EDTA chelation therapy. Meltzer et al. in

1961[25] showed that with 81 patients, 20 occurrences of mild hypocalcemia presented

Page 60: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

42

after EDTA chelation therapy (2000 infusions given on alternate days over 2 years).

Guldager et al. in 1993 [14] conducted a randomized controlled trial with 15 women and

39 men (aged 41 to 81 years, 20 intravenous infusion, 5-9 weeks) and showed a decrease

in serum calcium after EDTA chelation therapy.

Some other side effects include bone loss [14, 26]. Results for bone loss are non-

consistent. Rudolph et al. in 1988 [26] conducted a case study with 61 patients and

showed that EDTA chelation therapy (3 g EDTA, 20 intravenous infusion, 5-9 weeks)

had no effect on bone density and may even enhance bone growth in patients with

osteoporosis. On the contrary, Guldager et al. in 1993 [14] did a randomized controlled

trial with total 54 patients (15 women and 39 men, aged 41 to 81 years) with intermittent

claudication and showed that EDTA chelation therapy (3 g EDTA, 20 intravenous

infusion, 5-9 weeks) increased bone resorption and was accompanied by bone loss.

Page 61: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

43

Table 2.8. Side effects of EDTA chelation therapy.

Side effect Sample size Treatment

protocol

Results References

Renal toxicity 5; human NA Two instances of renal

tubular vacuolization

following administration of

sodium-EDTA have been

observed

Holland(1953)

[18]

NA NA Vacuolization occurred;

part of the chelate enters the

cell via pinocytosis

Schwartz(1966)

[19]

NA; rats NA Tubular vacuolization

produced in the rat by

intraperitoneal injection of

EDTA daily for 10 days;

Doolan(1967) [20]

SD rats, n=5

in each group

1000 mg/kg

disodium Ca-

EDTA, jugular

vein injection;

EDTA-induced

vacuologenesis is a

reflection of the induction of

pinocytosis by the chelate

Schwartz(1970)

[21]

383 patients;

3 g EDTA, 10

intravenous

infusion, 5 days

between infusions

EDTA infusion is not

nephrotoxic

McDonagh(1982)

[22]

NA NA Renal failure accompanied

with underlying renal

disease

Oliver(1984)[23]

Hypocalcemia NA NA Transient Hypocalcemia Spencer(1956)[24]

81 patients 2000 infusions

given on alternate

days over 2 years

20 occurrences of mild

Hypocalcemia

Meltzer(1961)[25]

Total 54

patients;

3 g EDTA, 20

intravenous

infusion, 5-9

weeks

Decreased in serum calcium Guldager(1993)

[14]

Bone loss 61 patients 3 g EDTA;

intravenous

infusion;

3 months

No effect to bone density;

Mayt enhance bone growth

in patients with osteoporosis

Rudolph(1988)

[26]

Total 54

patients;

with

intermittent

claudication

3 g EDTA, 20

intravenous

infusion, 5-9

weeks

Increased bone resorption;

Accompanied by bone loss

Guldager(1993)

[14]

NA: not available;

Page 62: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

44

2.5.3 Current opinions of EDTA chelation therapy for cardiovascular disease

Current opinions of EDTA chelation therapy for cardiovascular disease are still

quite disputed (Table 2.9). There are many positive opinions from proponents [17, 141,

168, 172] but there are many negative opinions from opponents [173-176]. Proponents

stated that “EDTA chelation can significantly decrease atheromatous plaque” [168],

“modestly reduced the risk of adverse cardiovascular outcomes” [17]. On the contrary,

opponents stated that “It was not useful for CAD or PVD” [173], “Chelation therapy is

not recommended for the treatment of chronic angina or atherosclerotic cardiovascular

disease and may be harmful because of its potential to cause hypocalcaemia” [174]. Most

proponents highlight therapeutics effects of EDTA chelation therapy in aspects of

cardiovascular outcomes but whether or not it reverses calcification is not clear.

EDTA treatment to treat hypercalcemia is FDA approved but EDTA drugs for

purpose of chelation is not approved by FDA. FDA has not received statistically

significant clinical data sufficient to support either the safety or efficacy of EDTA drugs

when they are used for these "chelation" purposes [140].

Page 63: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

45

Table 2.9. Current opinions of EDTA chelation therapy for cardiovascular disease.

References Organization Statements

Negative opinions from opponents

Soffer(1964)

[173]

Editor-in-Chief of both

Chest and Archives of

Internal Medicine

“It was not useful for CAD or PVD.”

Fraker(2007)

[174]

Cardiologist and professor

of clinical medicine at The

Ohio State University

Medical Center

“Chelation therapy (intravenous infusions of

ethylenediamine tetra-acetic acid or EDTA) is not

recommended for the treatment of chronic angina or

atherosclerotic cardiovascular disease and may be

harmful because of its potential to cause

hypocalcaemia.”

Jackson(2008)

[175]

Editor of International

Journal of Clinical Practice

“Chelation therapy is TACT-less, one wonders why it

is still widely used, given the lack of evidence of

benefit and potential to do harm (financial gain seems

the most likely reason).”

Atwood(2008)

[176]

Assistant Clinical Professor

at the Tufts University

School of Medicine

“We conclude that the TACT is unethical, dangerous,

pointless, and wasteful. It should be abandoned.”

“Chelation for atherosclerosis was condemned by the

Medical Letter, the American Heart Association

(AHA), and the American College of Physicians, the

American ....”

Positive opinions from proponents

Rudolph(1991)

[168]

Associate Professor and

Chairman in Biochemistry

Department of Texas

College of Osteopathic

Medicine

“EDTA chelation can significantly decrease

atheromatous plaque and the chance of cerebral

infraction.”

Cranton(2001)

[172]

President, American College

for Advancement in

Medicine, 1991-1992.

Editor-in-Chief, Journal for

Advancement in Medicine,

1986 to 1989.

"This therapy has been proven effective over and over

again in clinical practice."

"More than one million patients have received more

than twenty million infusions with no serious or lasting

adverse effects."

Lamas(2006,.

2013) [17, 141]

Chief, Division of

Cardiology at Mount Sinai

Medical Center, Columbia

University;

“EDTA chelation therapy meets evidence-based

medicine.”

“Among stable patients with a history of Myocardial

infarction (MI), use of an intravenous chelation

regimen with disodium EDTA, compared with placebo,

modestly reduced the risk of adverse cardiovascular

outcomes.”

CAD: coronary artery disease; MI: myocardial infarction; PVD: peripheral vascular disease; TACT: Trial

to Assess Chelation Therapy

Page 64: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

46

2.6 Targeted EDTA chelation therapy

2.6.1 Local drug delivery in the vasculature

MAC occurs heterogeneously along vascular tree. Thus local drug delivery would

a good option for treating MAC. Ideally local drug delivery would: 1) maximize drug’s

therapeutic effects and also 2) minimize undesired systemic side effects.

Three possible approaches to delivers drug locally to vasculature can be classified

as: 1) intra-luminal delivery; 2) peri-adventitial delivery; and 3) Novel sustained release

vehicles as showed in Figure 2.9, Figure 2.10, and Figure 2.11.

Some current available intra-luminal delivery devices include: double balloon

catheter (Figure 2.9A) and periadventitial delivery using needle injection catheter

(Figure 2.9B). Jo̸rgensen et al. in 1989 did a study with 6 consecutive patients

undergoing percutaneous transluminal angioplasty (PTA) for superficial femoral artery

occlusion, with a 7-French double-balloon catheter. Recombined human tissue-type

plasminogen activator (rt-PA) and heparin were then infused into the enclosed space for

30 minutes, followed by intravenous heparin for 24 hours. Results showed that at 10 and

30 days all 6 patients had evidence of recanalisation and remission of symptoms [177].

Needle injection catheter used fine needles that penetrate the media to deliver drugs.

Although it is invasive, a study done by Ikeno et al. in 2004 with 17 swine by delivering

Oregon green-labeled paclitaxel (OGP) and tacrolimus with this method showed limited

injury and trauma and also had retention of drugs up to 96 hours [178].

Page 65: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

47

Figure 2.9. Intra-luminal delivery devices. A: Double balloon catheter [144]; B:

Periadventitial delivery using needle injection catheter [178].

Several animal studies showed successful peri-adventitial delivery of therapeutics

in local vasculature. Peri-adventital delivery methods include: Drug eluting polymer wrap

(Figure 2.10A), and polyvinyl alcohol (PVA) foam sutured to the artery (Figure 2.10B).

Villa et al. in 1994 did a rat study using a silicone polymers wrapped around the common

carotid arteries for 3 weeks, and results showed that periadventitial local delivery of

dexamethasone markedly inhibits neointimal proliferation after balloon vascular injury

[179]. Researches also have used PVA foams loaded with drugs for peri-adventitial

delivery. Sho et al. in 2004 used a mini-osmotic pump loaded with doxycycline into

PVA foam in order to provide a continuous infusion via periaortic delivery system

(PDS) and showed that PDS (1.5 mg/kg/day) and subcutaneous (60 mg/kg/day) delivery

methods caused comparable reductions in abdominal aortic aneurysm (AAA) diameter

Page 66: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

48

during the period of 14 days. PDS rats gained more weight during the postoperative

period, possibly as a result of reduced serum drug levels and systemic toxicity [56].

Figure 2.10. Peri-adventital delivery methods. A: Drug eluting polymer wrap [179]; B:

PVA foam sutured to the artery [56].

Above mentioned delivery methods, intra-luminal and peri-adventitial delivery of

drugs to the site of pathology are all one time single application. However, sustained

release of drugs might be an attractive option to optimize drug’s therapeutic effects.

Some examples are discussed below.

Ogata et al. in 2010 developed a doxycycline loaded controlled release

biodegradable fiber (DCRBF) for local administration in AAA tissue (Figure 2.11A, B

and C).These doxycycline incorporated poly-lactic acid fibers showed a controlled in-

vitro drug release of 28 days and in-vivo release of up to 84 days [180].

Page 67: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

49

Franck et al. in 1998 successfully created hydrocortisone loaded microspheres and

delivered them via perforated balloon catheters into rabbit arteries after balloon

angioplasty [181]. Westedt et al. also formulated nanoparticles that were infused coupled

with porous balloon catheter (Figure 2.11D) [182].

Figure 2.11. Novel sustained release vehicles. A: gross appearance of DCBRF; B and

C:TEM images of DCBRF[180]. D: TEM showing the nanoparticles eluting out of the

porous balloon [182].

All above mentioned local delivery methods require surgical manipulation which

would be quite invasive for patients. Also for several vascular calcification diseases, the

diseased aorta is spread heterogeneously along vascular tree. Thus an ideal delivery

method should be able to be applied systemically and targeted to various sites of

calcification. One possible way is using targeted albumin nanoparticles as discussed

below.

Page 68: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

50

2.6.2 Albumin nanoparticles as drug carriers for different application

Albumin, a versatile protein carrier for drug delivery, is nontoxic, non-

immunogenic, biocompatible and biodegradable [183]. Therefore, it is ideal material to

fabricate nanoparticles for drug delivery.

Types of albumin used as drug carrier in nanoparticles include: Ovalbumin,

Bovine serum albumin (BSA) and Human serum albumin (HSA). Bovine serum albumin,

with a molecular weight of 69 KDa and an isoelectric point of 4.7 in water (at 25 °C), is

widely used for drug delivery because of its medical importance, abundance, low cost,

ease of purification, unusual ligand-binding properties, and its wide acceptance in the

pharmaceutical industry [184]. BSA could be replaced by HSA in order to circumvent a

possible immunologic response in vivo. HSA has preferential uptake in tumor and

inflamed tissue and its ready availability, biodegradability and lack of toxicity make it an

ideal candidate for drug delivery [185].

Methods to prepare albumin nanoparticles include: desolvation [186],

emulsification [187], thermal gelation [188] and recently nano spray drying [189], nab-

technology [190] and self-assembly [191]. In desolvation process, nanoparticles are

obtained by a continuous dropwise addition of ethanol to an aqueous solution of albumin

(pH 5.5) under continuous stirring until the solution becomes turbid. During the addition

of ethanol into the solution, albumin is phase separated due to its diminished water-

solubility. Coacervates are hardened by crosslinking with glutaraldehyde where the

amino moieties in lysine residues and arginine moieties in guanidine side chains of

Page 69: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

51

albumin are solidified by a condensation reaction with the aldehyde-group of

glutaraldehyde [192].

Many in vivo studies have shown BSA to be a suitable carrier to target gamma-

interferon (IFN-γ) to macrophages and thus potentiating their therapeutic activity [193].

In a platelet aggregometric study, Das et al. clearly showed that the amount of aspirin

diffused from aspirin-loaded BSA nanoparticles at the end of 48 h was sufficient to

prevent platelet aggregation [194].

Many surface-modified albumin nanoparticles using antibody have been

investigated for better targeting. It has been shown that many malignancies like colorectal,

head, neck, non-small cell lung ovarian, breast and prostate cancers, as well as glioma,

show an overexpression of the epidermal growth factor receptor (EGFR) on their surfaces.

Thus, cetuximab, a humanized IgG1 monoclonal antibody (mAb) targets EGFR and was

approved for the treatment of colorectal cancer by FDA in 2004 [195]. Therefore,

cetuximab-modified HSA nanoparticles are a promising carrier system for drug transport.

A specific accumulation targeting the EGFR could be shown in EGFR-expressing colon

carcinoma cells [195]. Besides, DI17E6, a monoclonal antibody directed against αv-

integrins, was covalently coupled to HSA nanoparticles loaded with doxorubicin [196].

The thiolated antibody was coupled with sulfhydryl-reactive nanoparticle suspension to

achieve a covalent linkage between antibody and the nanoparticle system. The target-

specific nanoparticles specifically targeted αvβ3 integrin positive melanoma cells

showing increased cytotoxic activity compared to the free drug [196]. Since many

Page 70: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

52

antibody coated albumin nanoparticles have been approve by FDA and are in clinical use,

our strategy to use anti-elastin antibody coated albumin nanoparticles would be also have

great clinical relevance.

2.6.3 Targeted EDTA chelation therapy using EDTA loaded albumin

nanoparticles

Previous attempts at systemic chelation therapy with EDTA to reverse

calcification required high dosage and had many side effects such as hypocalcemia, renal

toxicity and bone loss [14, 18, 119]. We propose that targeted EDTA chelation therapy

might be a promising way to address issues accompanied with systemic chelation therapy.

Nanoparticle-based targeted drug delivery could lower dosage of EDTA required

and have many other advantages such as improved bioavailability and sustained release

of drugs [29]. We propose that nanoparticle protein engineering therapy using targeted

EDTA loaded albumin NPs with elastin antibody offers potential solutions to reverse

vascular calcification and also circumvent possible side effects from systemic chelation

therapy by reducing the high dosage that systemic infusion requires. More specific data

has been discussed in Chapter 7.

Page 71: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

53

CHAPTER 3: PROJECT RATIONALE

3.1 Project objective and aims

Based on the literature reviewed earlier, the main objectives of this project are:

a) Understand mechanisms of elastin specific medial arterial calcification

(MAC), specifically cause-and-effect relationship between the elastin-

specific MAC and osteoblast-like differentiation of vascular smooth

muscle cells (VSMCs).

b) Evaluate the efficacy of several chelating agents to reverse elastin specific

MAC.

c) Evaluate the therapeutic effects of systemic EDTA chelation therapy in the

aspect of reversal of calcification and possible side effects.

d) Develop a targeted delivery system based on EDTA loaded nanoparticles

to reverse elastin specific MAC at low doses and to circumvent systemic

side effects.

3.2 Specific aims and rationale

Specific Aim 1:

Determine cause-and-effect relationship between the elastin-specific MAC and

osteoblast-like differentiation of vascular smooth muscle cells (VSMCs).

Rationale: Osteoblast-like differentiation of vascular smooth muscle cells

(VSMCs) has been found in vascular calcification sites in animal studies as well as in

Page 72: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

54

clinical retrospective studies; however, whether this osteogenesis plays a major role in

the initiation of vascular calcification is unclear. We hypothesize that the initial

deposition of hydroxyapatite-like mineral in MAC occurs on degraded elastin first and

that causes osteogenic transformation of VSMCs. Thus, we wanted to test if rat aortic

smooth muscle cells (RASMCs) cultured on hydroxyapatite crystals and calcified aortic

elastin would transform to osteoblast like cells. Another interesting question is upon

removal of calcified conditions, whether or not differentiated cells would have the ability

to revert back to SMC-like cell behavior. This is important as we can then envision

demineralizing strategies to remove mineral from arteries and bring homeostasis to the

arterial structures.

Specific Aim 2:

Evaluate the efficacy of chelating agents to reverse elastin specific MAC.

Rationale: Chelation therapy has been touted as an effective treatment to reverse

vascular calcification; however, it has been controversial with opposing views [8-10].

Most of the previous work is focused on atherosclerotic intimal calcification and

unfortunately, the efficacy of various chelating agents in reversing elastin specific

calcification from the peripheral vascular tissue is not studied very well either in vitro or

in animal experiments. We want to assess the chelating ability of EDTA, DTPA, and STS

on removal of calcium from hydroxyapatite (HA) powder, calcified aortic elastin, and

calcified human aorta in vitro. We further want to assess whether or not that local

periadventitial delivery of EDTA loaded nanoparticles could reverse elastin specific

Page 73: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

55

calcification in the aorta. This research will allow us to select the best chelating agent for

reversal of MAC in vivo.

Specific Aim 3:

Evaluate therapeutic effects of systemic EDTA chelation therapy for reversal of

elastin specific MAC and its possible systemic side effects.

Rationale: At present whether or not EDTA chelation therapy reverses vascular

calcification is still quite disputed [9]. Currently, most of the EDTA chelation therapies

studied were using EDTA by subcutaneous infusion, or systemic application of EDTA

solution at very high dose (around 40 ~ 50 mg/kg) [11-17]. Many side effects have been

found with such a high systemic dose of EDTA chelation therapy including: renal

toxicity [18-23] , hypocalcemia [14, 24, 25], and bone loss [14, 26]. But results are quite

varied for side effects. We want to investigate whether or not systemic EDTA chelation

therapy could reverse local aortic calcification in an animal model of MAC and test

possible side effects to serum Ca, urine Ca, and bone loss. This would provide an

important pre-clinical animal study investigating EDTA’s therapeutic as well as potential

side effects in the aspect of reversal of MAC.

Specific Aim 4:

Develop a targeted delivery system for EDTA to reverse elastin specific MAC

and to avoid side effects associated with systemic therapy.

Page 74: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

56

Rationale: Systemic chelation therapy using EDTA or sodium thiosulfate to

reverse calcification requires high dosage and has side effects such as hypocalcemia,

bone loss, and renal toxicity [14, 18, 119]. If we can target EDTA to the local vascular

calcification sites, we could lower the dosage of EDTA and can also improve efficacy

and reduce side effects [29]. We have recently shown that the elastin antibody coated

nanoparticles (NPs) can be targeted to vascular calcification sites [30]. Thus, in this study

we want to test if albumin NPs may be a good drug carrier to load EDTA. By surface

coating of anti-elastin antibody, these elastin antibodies coated and EDTA loaded

albumin NPs could be a promising nanoparticle protein engineering therapy to reverse

elastin-specific medial calcification and reduce side effects associated with systemic

EDTA chelation therapy.

3.3 Clinical significance

Cardiovascular diseases (CVD) are the leading cause of death worldwide.

Pathological vascular calcification has been observed in many CVD and is recognized as

a major risk factor for mortality and morbidity in patients with diabetes and chronic

kidney disease. The mechanisms of medial arterial calcification (MAC) are not

understood well and currently there is no available alternative to surgical repair for

reversal of vascular calcification [6, 7]. One possible approach is EDTA chelation

therapy that has been tested in animals and in clinical trials. However, clinical trials did

not prove statistically significant improvement in cardiovascular function and thus such

therapies are not approved by FDA. Moreover, such chelation therapies have shown

Page 75: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

57

many side effects such: renal toxicity [18-23] , hypocalcemia [14, 24, 25], and bone loss

[14, 26].

In this research we wanted to shed light on the mechanisms of elastin specific

MAC, specifically cause-and-effect relationship between the elastin-specific MAC and

osteoblast-like differentiation of vascular smooth muscle cells (VSMCs). This research

would pave the way for mechanistic based therapies for vascular calcification.

Secondly, although EDTA chelation therapy has been tried for atherosclerotic

calcification, no research has looked at its effectiveness in reversing elastin-specific

MAC. If successful, such therapies would help millions of patients with end stage renal

disease and diabetes where MAC is prevalent. The literature also suggests that chelation

therapies are not approved by FDA for vascular calcification due to its systemic side

effects. No research is performed so far to see if EDTA chelation can be targeted to

vascular calcification sites by some sort of drug delivery approaches. Our research

presented in this thesis could pave the way to target chelation therapies vascular

calcification sites and eliminate systemic side effects.

Page 76: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

58

CHAPTER 4: MECHANISMS OF ELASTIN SPECIFIC MEDIAL ARTERIAL

CALCIFICATION

(This work has been published in

Experimental Cell Research 2014; 323:198-208)

4.1 Introduction

There are two distinctive types of vascular calcification: intimal atherosclerotic

plaque calcification and medial elastin-specific arterial calcification (MAC). Intimal

calcification occurs in the context of atherosclerosis, associated with lipids, macrophages,

and vascular smooth muscle cells; whereas, medial calcification can exist independently

of atherosclerosis and is associated with elastin and vascular smooth muscle cells[3]. In

this study we mainly focused on the mechanisms of MAC. It has been accepted for

decades that the pathology of vascular calcification resembles physiological bone

mineralization in that it shows the presence of bone proteins and osteo-chondrogenic cells

[197, 198]. However, mere presence of osteogenic cells and bone protein expression in

the calcified arterial tissue does not warrant the role of osteogenesis in the initiation of

elastin calcification. The scientific knowledge gap still exists about the cause-and-effect

relationship between the elastin-specific medial calcification and osteoblast-like

differentiation of vascular smooth muscle cells (VSMCs). It is still unclear if there exists

a chronological occurrence between the osteoblast-like differentiation of VSMCs and

passive deposition of hydroxyapatite on elastin, or if both processes occur simultaneously

causing the eventual calcification of arteries.

Page 77: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

59

Based on our previous in vivo data [199, 200], we hypothesize that the initial

mineral deposition on elastin precedes the osteoblast-like differentiation of VSMCs,

which is a pathological response to elastin degradation and early arterial calcification. To

test this hypothesis, we exposed rat aortic smooth muscle cells (RASMCs) to calcified

conditions, by culturing them on hydroxyapatite and calcified elastin. We show when

exposed to a calcific environment, healthy RASMCs transform to osteoblast-like cells;

removal of calcific environment restores the native phenotype of SMCs.

4.2 Materials and methods

Cell culture and treatment

Primary rat aortic smooth muscle cells (RASMCs) were isolated from rat aorta

according to published protocol[201]. Passage numbers 5 to 8 were used for all

experiments. Cells were cultured in 60 mm tissue culture petri dish (2×106 cells/well) in

Dulbecco's Modified Eagle Medium (Cellgro-Mediatech, Manassas, VA), containing

10% fetal bovine serum (Cellgro-Mediatech, Manassas, VA), 100 units/ml penicillin and

100 units/ml streptomycin (Cellgro-Mediatech, Manassas, VA) in a humidifier incubator

(Innova® CO-170) at 37 ℃, with 5% CO2. Media was replenished every 3 days.

Hydroxyapatite powder (average size <200 um) was purchased from Aldrich Chemical

Company (Milwaukee, WI). One mg of hydroxyapatite was coated onto 60 mm tissue

culture Petri dish. Here the coating process was achieved by first placing a

hydroxyapatite suspension in sterile phosphate-buffered saline (PBS) on a Petri dish and

then drying it overnight under sterile cell culture hood. For control group, the RASMCs

Page 78: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

60

were cultured in a Petri dish without hydroxyapatite coating. To better mimic the

calcified pathological situation, RASMCs were also cultured in a Petri dish with a coating

of calcified porcine aortic elastin fibers (10 mg per well) while a coating with non-

calcified elastin served as a control. The calcified porcine aortic elastin fibers were

prepared in vitro based on a modified protocol[202] and the average size for both the

pure and calcified elastin fiber were less than 200 µm (passed through 200 µm sieve).

Briefly purified elastin was suspended in a calcifying solution (Tris-HCl buffer PH7.4;

KCl 55 mmol/l; KH2PO4 35 mmol/l; CaCl2 35 mmol/l) for 7 days with shaking at 37℃.

Poorly crystalline hydroxyapatite was deposited on elastic fibers. The calcium deposition

was characterized by alizarin red stain and scanning electron microscope (SEM, Hitachi's

SU6600) with energy dispersive X-ray spectrometer (EDX) and quantified by atomic

absorption spectroscopy (Perkin-Elmer Model 3030, Norwalk, CT). Cells were grown on

top of calcified matrix for 1 day and 7 days. To study the RASMCs’ fate after removing

calcified conditions, the cells were first cultured for 7 days on calcified matrix

(specifically both hydroxyapatite and calcified elastin) followed by isolation and

placement on a 60 mm tissue culture Petri dish without calcified matrix for an additional

7 days, or 14 days total as referred to below (n=12 per group total, 3 for RNA isolation, 3

for protein isolation, 3 for cell immunofluorescence and another 3 for staining for

alkaline phosphatase). The experiments were repeated twice.

Gene expression

Page 79: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

61

At each time point, cell monolayers were scraped and homogenized (n=3 per

group) using a homogenizer (PowerGen Model 125 Homogenizer, Fisher Scientific,

Atlanta, GA). The total RNA from the cells was isolated using the RNeasy Mini Kit

(Qiagen, MD). The concentration of the extracted RNA was quantified by UV

spectroscopy using Take3 Micro-Volume Plates from BioTek (Winooski, VT). One

microgram of RNA was reverse transcribed to cDNA by High Capacity cDNA Reverse

Transcription Kit (Applied Biosystems, Foster City, CA) using a Mastercycler gradient

(Eppendorf Scientific, Inc., Westbury, NY). The cDNA samples were further amplified

on a Rotor gene 3000 thermal cycler (Corbett Research, Mortlake, NSW, Australia) for

real - time polymerase chain reaction (PCR). The primers sequences for β-2microglobulin

(β2-MG)[200], alpha smooth muscle actin (SMA)[203], myosin heavy chain

(MHC)[204], aggrecan[205], collagen type II alpha 1(Col2a1)[206], alkaline phosphatase

(ALP)[200], Runt-related transcription factor 2 (RUNX-2)[200] and osteocalcin[200] are

listed in Table 4.1. Gene expression in each sample was normalized to the expression of

β2-MG as housekeeping gene and compared with control samples (for the hydroxyapatite

study group, the control was the cells grown on standard culture plates while for the

calcified elastin group, the control was the cells grown on non-calcified elastin coated

culture plates) using the 2-ΔΔC

T method[207] as follows: ΔΔC

T= (CT target gene-CT β-2

MG) study – (CT target gene-CT β-2 MG) control.

Page 80: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

62

Table 4.1. PCR primers used in the RT-PCR study.

Name References Forward primer Reverse primer

β2 MG Lee et al[200]. CGTGATCTTTCTGGTGCTTGTC ACGTAGCAGTTGAGGAAGTTGG

SMA Simionescu et al[203] ACTGGGACGACATGGAAAAG CATACATGGCAGGGACATTG

MHC Low et al[204]. AAGCAGCTCAAGAGGCAG AAGGAACAAATGAAGCCTCGTT

Aggrecan Agung et al[205]. TAGAGAAGAAGAGGGGTTAGG AGCAGTAGGAGCCAGGGTTAT

Clo2a1 Zhang et al[206]. TCCTAAGGGTGCCAATGGTGA AGGACCAACTTTGCCTTGAGGAC

OCN Lee et al[200]. TATGGCACCACCGTTTAGGG CTGTGCCGTCCATACTTTCG

ALP Lee et al[200]. TCCCAAAGGCTTCTTCTTGC ATGGCCTCATCCATCTCCAC

RUNX2 Lee et al[200]. CAACCACAGAACCACAAGTGC CACTGACTCGGTTGGTCTCG

Immunofluorescence

The cells were fixed in 4% paraformaldehyde (Affymetrix, Cleveland, OH) for 15

minutes at room temperature, treated with 1% bovine serum albumin (Sigma, St. Louis,

MO)/0.2% Triton X-100 (MP Biomedicals, Solon, OH)/PBS for 1 hour at room

temperature to block non-specific binding. The primary antibodies used include: mouse

monoclonal Anti- SMA at 1:200 dilutions (Sigma, St. Louis, MO), mouse monoclonal

[1C10] to smooth muscle Myosin heavy chain I at 1:200 dilution (Abcam, Cambridge,

MA) and mouse anti-collagen type I at 5 µg/ml (Developmental Studies Hybridoma

Bank, Iowa City, IA). After overnight incubation at 4 °C, cells were stained for 2 hours at

room temperature in the dark with AlexaFlour 594 donkey anti-mouse IgG secondary

antibody (Molecular Probes, Eugene, OR) diluted to 10 µg/ml. All well plates were

mounted in VECTASHIELD HardSet Mounting Medium (VECTOR LABORATORIES,

INC, Burlingame, CA) with DAPI (4', 6-diamidino-2-phenylindole dihydrochloride) blue

fluorescent nuclear stain (Molecular Probes, Eugene, OR) and examined by fluorescence

microscopy.

Page 81: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

63

Protein isolation

Cell monolayers were washed once with PBS and isolated in a mammalian

extraction buffer. To prepare the buffer, 1 tablet of protease inhibitor cocktail (Roche

Diagnostics, Indianapolis, IN) was added to 10ml of SoluLyse-M™ Mammalian Protein

Extraction Reagent (Genlantis, San diego, CA). SoluLyse-M™ reagent (500 µl) was

added into the 60 mm tissue culture petri dishes. Cells were incubated for 10 minutes at

25 ℃ with gentle rotation and then homogenized and centrifuged at 14,000 X g for 5

minutes. The supernatant was collected (preserved in -20℃ freezer) and assayed for

different proteins of interest. The total cellular protein was quantified by Pierce® BCA

protein assay kit (Thermo scientific, Rockford, IL).

Western blotting

Ten micrograms of protein from each sample and molecular weight standards

(Precision Plus Protein™ Kaleidoscope Standards, Bio Rad Life Science, Hercules, CA),

were loaded in duplicate on 10% sodium dodecyl sulfate-polyacrylamide gel

electrophoresis gels. After electrophoresis, the proteins were electro-transferred to

Immobilon-P membranes (Millipore Corporation, Bedford, MA). Membranes were

blocked in 2% nonfat dry milk (LabScientific, Inc., New Jersey) for 1 hour at room

temperature and then probed with primary antibody overnight at 4°C. Primary antibodies

used include: mouse monoclonal Anti- SMA at 1:1000 dilution (Sigma, St. Louis, MO),

mouse monoclonal [1C10] to smooth muscle Myosin heavy chain I at 1:1000 dilution

(Abcam, Cambridge, MA) and rabbit monoclonal [EP2978Y] to beta 2 Microglobulin

Page 82: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

64

(Abcam, Cambridge, MA). The proteins were detected by enhanced chemiluminescence

according to the manufacturer’s recommendations (Roche, Indianapolis, IN) and

analyzed by densitometry using Image J software. The optical densities of the bands were

reported as relative density units (RDU) comparing to the control groups. Beta 2

Microglobulin antibodies were used as the loading control and the target protein’s band

densities were first normalized by beta 2 Microglobulin.

ALP assay – colorimetric quantification and cell stain

Cell lysates were analyzed for alkaline phosphatase (ALP) using p-nitrophenyl

phosphate (PNPP) as a substrate and diethanolamine buffer. Two PNPP tablets (Thermo

scientific, Rockford, IL) were dissolved in 10 ml diethanolamine substrate buffer (Pierce,

Rockford, IL). PNPP solution (200 µl) and protein samples (40 µl) were added into each

well of the 96-well plate. The plate was incubated at room temperature for 30 minutes or

until sufficient color developed. The reaction was stopped by adding 50 µl of 2N NaOH

to each well. The absorbance was measured at 405 nm. Alkaline phosphatase was

calculated using a p-nitrophenol (MP Biomedicals Inc, Solon, Ohio) standard curve and

was normalized to the total protein content. Additionally, cells in culture were stained to

corroborate the expression of ALP by the cells. Cells were stained with premixed

BCIP/NBT solution (Sigma Aldrich, St. Louis, MO). The premixed BCIP/NBT solution

contains 0.48 mM NBT (nitro blue tetrazolium), 0.56 mM BCIP (5-bromo-4-chloro-3-

indolyl phosphate), 10 mM Tris HCl, 59.3 mM MgCl2, and pH ~9.2. Cells were

Page 83: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

65

incubated in the staining solution in the dark for 1 hour at room temperature and then

washed.

Osteocalcin Assay- Enzyme-linked immunosorbent assay (ELISA)

The total cellular protein samples were analyzed for osteocalcin using a Rat

Osteocalcin ELISA Kit (Biomedical Technologies Inc., Stoughton, MA) following the

manufacturer’s instructions. 25 µl of total protein sample, rat osteocalcin serial

standards(1.0, 2.5, 5.0, 10 and 20 ng/ml) and rat serum controls were pipetted into

osteocalcin antibody coated well plate strips followed by 100 µl of osteocalcin

antiserum(second antibody, goat polyclonal) at 37 °C for 2.5 hours. Donkey anti-goat

IgG peroxidase conjugate was used as secondary antibody and 3, 3′, 5, 5′-

Tetramethylbenzidine (TMB) was used as the peroxidase substrate. Levels of osteocalcin

were calculated by rat osteocalcin standard curve and then normalized to total protein

concentration.

Statistical Analysis

Results are expressed as means ± standard error of the mean (SEM). Statistical

analyses of the data were performed using single-factor analysis of variance. Differences

between means were determined using the least significant difference with an α value of

0.05. Asterisks in figures denote statistical significance (P < 0.05) for each group

compared with controls.

Page 84: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

66

4.3 Results

Characterization of calcified elastin

Calcified elastin was prepared in vitro and the calcium level in the calcified

elastin fiber was detected as 15 ± 2 µg/mg using atomic absorption spectroscopy. The

calcium deposition was evenly distributed over the porcine elastin fiber as seen by

alizarin red stain, which stains calcium red while no staining was seen in the control pure

elastin (Figure 4.1A). The morphology of calcified elastin, as studied by scanning

electron microscopy, showed globular calcific deposits. These were confirmed as calcium

phosphates by Energy-dispersive X-ray spectroscopy (Figure 4.1B and 4.1C).

Page 85: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

67

Figure 4.1. Characterization for calcified elastin. A: Alizarin red staining for pure elastin

(PE, left) and calcified elastin (CE, right). Calcium was stained red. Scale bar: 50µm. B:

Scanning electron microscope (SEM) image for pure elastin (PE, left) and calcified

elastin (CE, right). C: Energy-dispersive X-ray spectroscopy (EDX) pseudo colored

image for Ca and P element distribution (Ca purple and P red). Mineral deposition is

labeled with red arrows.

Page 86: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

68

RASMCs lose smooth muscle lineage markers under calcified conditions

We analyzed gene and protein level expression of two typical smooth muscle

lineage markers: alpha smooth muscle actin (SMA)[208] and myosin heavy chain

(MHC)[209] in two different simulated calcified conditions: (1) pure hydroxyapatite; (2)

calcified elastin. These markers were studied at two time points (day 1 and day 7) to see

the changes compared to the control groups. Cellular localization based on

immunofluorescence stain of SMA showed that both hydroxyapatite and calcified elastin

groups had similar SMA expression at day 1 as controls; however, at day 7 they exhibited

very low expression of SMA protein (Figure 4.2A). Similarly, immunofluorescence stain

for MHC also showed little change at day 1 but a significant decrease was observed at

day 7 (Figure 4.2B).

Page 87: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

69

Page 88: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

70

Figure 4.2. Expression of SMA and MHC in RASMCs cultured on hydroxyapatite and

calcified elastin at day 1 and day 7. A and B: Immunofluorescence stains of SMA and

MHC respectively (red fluorescence). Nuclei were stained blue by DAPI. Scale bar:

100µm. Abbreviations: HA-hydroxyapatite; PE: pure elastin; CE: calcified elastin. D1:

day 1; D7: day 7. C and D: SMA and MHC gene expression respectively as measured by

RT-PCR. E: protein expression measured by western blot followed by densitometry as

relative density units (RDU) and expressed as relative percentage ratio comparing to

control groups (For HA treatment group the control was normal media while for CE

treatment group the control was PE). The band density was first normalized by loading

control protein- β2 MG. Results are presented as mean ± SEM (* p<0.05).

Furthermore, both hydroxyapatite and calcified elastin treated groups showed

decrease in the gene expression (quantified by RT-PCR) and protein expression (detected

by western blot) of SMA and MHC at day 7. Compared to respective control groups,

SMA gene expression decreased by 48 ± 18% in hydroxyapatite groups and 53 ± 5% in

calcified elastin groups (Figure 4.2C). Similarly, MHC gene expression decreased by 64

± 19% in hydroxyapatite groups and 60 ± 13% in calcified elastin groups (Figure 4.2D).

Western blotting results confirmed the down-regulation of translation of both SMC

markers. Compared to respective control groups, the SMA protein expression decreased

by 56 ± 2 % in hydroxyapatite groups and 48 ± 2% in calcified elastin groups (Figure

4.2E). The MHC protein expression decreased by 52 ± 6% in hydroxyapatite groups and

53 ± 2% in calcified elastin groups (Figure 4.2E).

RASMCs develop an osteogenic/chondrogenic phenotype under calcified conditions

We then analyzed expression of three typical osteogenic markers: Runt-related

transcription factor 2 (RUNX2) [210, 211], Alkaline phosphatase (ALP) and osteocalcin

(OCN)[208, 209, 212, 213] and two typical chondrogenic markers: aggrecan and

Page 89: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

71

collagen type II alpha 1(Col2a1) to see whether RASMCs develop an

osteogenic/chondrogenic phenotype with the stimuli of hydroxyapatite and calcified

elastin. At day 1, we found significant upregulation of RUNX2 gene under mineralized

conditions, which is the earliest transcription factor involved in osteogenesis. No other

osteogenic markers were changed at day 1 as compared to controls (Figure 4.3A).

Compared to respective control groups, RUNX2 gene expression increased by 87 ± 43%

in hydroxyapatite groups and 116 ± 19% calcified elastin groups at day 7. Similarly, at

day 7, ALP gene expression increased by 41 ± 20% in hydroxyapatite groups and 188 ±

21% in calcified elastin groups while OCN gene expression increased by 111 ± 30% in

hydroxyapatite groups and 54 ± 21% in calcified elastin groups (Figure 4.3A).

Page 90: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

72

Page 91: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

73

Figure 4.3. Expression of osteogenic/chondrogenic markers in RASMCs cultured on

hydroxyapatite and calcified elastin elastin at day 1 and day 7. A: gene expression of

RUNX2, ALP and OCN. B: Histochemical staining of ALP activity in cells in four

groups (C-control, HA-hydroxyapatite, PE-pure elastin, CE-calcified elastin). Scale bar:

50µm. C: ALP protein quantification. D: OCN protein quantification. OCN was

determined by ELISA assay and expressed as ng osteocalcin /mg protein. The relative

protein expression for both ALP and OCN has been calculated compared to respective

control groups. E: Gene expression of chondrogenic markers – aggrecan. F: Gene

expression of chondrogenic markers – Col2a1. G: Immunofluorescence stains for

collagen type I (red fluorescence). Nuclei were stained blue by DAPI. Scale bar: 100µm.

Results are presented as mean ± SEM (* p<0.05).

Histochemical staining for ALP revealed intense purple coloration in both

hydroxyapatite and calcified elastin groups indicating greater ALP activity compared to

control groups (Figure 4.3B). Additionally, quantification of cellular ALP activity

showed an increase by 207 ± 35 % in hydroxyapatite groups and 72 ± 9 % in calcified

elastin groups at day 7 (Figure 4.3C). Osteocalcin protein expression increased by 97 ±

13 % in hydroxyapatite groups and 23 ± 12 % in calcified elastin groups respectively at

day 7 (Figure 4.3D). The ALP and OCN protein expression in calcified elastin groups

was much less elevated might because of relative lower concentration of hydroxyapatite

in elastin group, specifically one mg of of HA in hydroxyapatite versus only ~150 µg in

elastin group (10 mg of calcified elastin with mineral deposition of 15 ± 2 µg Ca /mg

tissue) (Figure 4.3C, 4.3D). It is also possible that HA crystal structure in two groups is

different and that can cause variation in relative expression.

For the chondrogenic markers, at day 7, compared to respective control groups,

Aggrecan gene expression increased by 125 ± 76% in hydroxyapatite groups and 179 ±

84% in calcified elastin groups (Figure 4.3E); Col2a1 gene expression increased by 613

Page 92: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

74

± 108% in hydroxyapatite groups and 370 ± 141% in calcified elastin groups (Figure

4.3F).

RASMCs show up-regulation of Collagen Type I under calcified conditions

Immunofluorescent detection of collagen type I showed an intense signal both for

hydroxyapatite and calcified elastin treated group at day 7 (Figure 4.3 G). The increase

of collagen type I expression under calcified conditions, indicated more synthetic

phenotype of RASMCs under mineralized conditions.

RASMCs restore their original phenotype after removal of calcified matrix

In an independent set of experiments, RASMCs were cultured on calcified matrix

for 7 days and then were cultured for another 7 days without the calcified matrix to study

cellular behavior after removing calcified matrix. After 14 days (7 days post reversal)

both hydroxyapatite and calcified elastin treated group showed dramatic restoration of

VSMCs markers (comparable to control groups: for the hydroxyapatite study group, the

control was the cells grown on standard culture plates while for the calcified elastin

group; the control was the cells grown on non-calcified elastin coated culture plates at

day 14) as detected by immunofluorescence (Figure 4.4A, 4.4B) and RT-PCR and

western blotting (Figure 4.4C, 4.4D). All the earlier mentioned osteogenic/chondrogenic

markers including RUNX2, ALP, OCN, aggrecan and col2a1 decreased gene expression

at day 14 and restored close to normal level (Figure 4.5A, 4.5B). The protein expression

of ALP and OCN also decreased comparing to day 7 although still remained elevated

Page 93: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

75

comparing to the normal control levels (Figure 4.5C). Histochemical staining for ALP

restored to normal level comparing to the controls (Figure 4.5D). Collagen type 1

expression also restored close to normal level (Figure 4.5E). These results indicate that

the phenotypic transition from smooth muscle cells to osteoblast-like cells is reversible

with reversal of calcification.

Page 94: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

76

Figure 4.4. Expression of SMA and MHC in RASMCs after removal of calcified matrix.

Cells were cultured on calcified matrix for 7 days, then removed and cultured in normal

cell culture plate for additional 7 days (total 14 days of culture) A: Immunofluorescence

stains of SMA (red fluorescence). B: Immunofluorescence stains of MHC (red

fluorescence). Nuclei were stained blue by DAPI. Scale bar: 100µm. Abbreviations: C-

control, HA-hydroxyapatite; PE: pure elastin; CE: calcified elastin. D14: day 14. C:

SMA and MHC gene expression measured by RT-PCR. D: protein expression measured

by western blot followed by densitometry as relative density units (RDU) and expressed

as relative percentage ratio comparing to control groups (For HA treatment group the

control was normal media while for CE treatment group the control was PE). The band

density was first normalized by loading control protein- β2 MG. Results are presented as

mean ± SEM (* p<0.05).

Page 95: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

77

Figure 4.5. Expression of osteogenic/chondrogenic markers in RASMCs after removal of

calcified matrix. Cells were cultured on calcified matrix for 7 days, then removed and

cultured in normal cell culture plate for additional 7 days (total 14 days of culture) A:

gene expression of RUNX2, ALP and OCN. B: gene expression of aggrecan and Col2a1.

C: protein quantification of ALP and OCN. D: Histochemical staining of ALP activity in

cells at in four groups(C, HA, PE, CE). Original magnification, 400X. Scale bar: 50µm.

E: Immunofluorescence stains for collagen type I (red fluorescence). Nuclei were stained

blue by DAPI. Scale bar: 100µm. Results are presented as mean ± SEM (* p<0.05).

4.4 Discussion

Our data shows that, in response to the calcified matrix, for both hydroxyapatite

crystals and calcified aortic porcine elastin, RASMCs lose their smooth muscle lineage

markers, specifically alpha smooth muscle actin (SMA) and myosin heavy chain (MHC).

In addition, RASMCs undergo chondroblast/osteoblast-like differentiation confirmed by

Page 96: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

78

an increase in expression of typical bone-markers such as Runt-related transcription

factor 2 (RUNX2), alkaline phosphatase (ALP), osteocalcin (OCN), aggrecan, and

collagen type II alpha 1(Col2a1). RASMCs also turn synthetic as indicated by increased

expression of collagen type I. Interestingly, this phenotypic transition was reversible and

RASMCs restored their original linage upon reversal of calcification.

Putative relationship between osteoblast-like transformation of VSMCs and

vascular calcification is still under debate and the cause-and-effect relationship between

these two processes is unclear. It has been reported that the vascular calcification process

can be caused by osteoblast-like differentiation of VSMCs[214]. Chronic kidney disease

(CKD) related vascular calcification has often been associated with increase serum Ca

and P levels[71]. Elevated phosphate conditions could cause osteogenic/chondrogenic

differentiation of VSMC [208, 215]. In rat models of renal failure, animals with severe

calcification showed the presence of chondrocyte-like cells. Mature cartilage tissue and

major chondrogenic factors were found in the calcified vessels[216]. Osteoblast-like

differentiation of VSMCs was also reported in the aortas of transgenic mice, ubiquitously

expressing Msx2 (encodes an osteoblast-specific transcription factor), that were fed with

a high-fat diet. High-fat diets led to vascular calcification in these transgenic mice but not

in their non-transgenic littermates[217]. In type-2 diabetes related medial calcification,

BMP-2/Mxs2/Wnt signaling has been established[218]. Studies in patients with type 2

diabetes showed that medial calcification is a cell-mediated process characterized by a

phenotypic change of VSMCs to osteoblast-like cells[219]. Based on these studies, it is

Page 97: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

79

thought that chondrogenic/osteogenic transformation of VSMCs is the earliest step in

vascular calcification.

Alternatively, there are many studies support that medial elastin-specific

calcification can occur due to degraded elastin without osteoblastic differentiation of

VSMCs. In early 1970s, Urry has shown that elastin has a specific calcium binding site

serving as the neutral nucleation site for calcium [220]. We previously showed that pure

aortic elastin when implanted subdermally in rats undergoes calcification with bone-

protein expression occurring only in the later stages of mineral propagation[200]. In

circulatory rat model, we confirmed that matrix metalloproteinases (MMPs) mediated

degradation of elastin, leads to elastic-lamina calcification in absence of cell-associated

calcification [199]. Price et al. have shown that in the presence of serum factors, arterial

elastin calcifies in vitro in absence of cells[221]. Furthermore, VSMCs have shown

osteoblast-like behavior in vitro when exposed to elastin peptides along with TGF-beta,

therefore suggesting a causative role of elastin degradation in VSMCs mediated

calcification [212]. Recently Murshed et al found in a MGP-deficient (Mgp−/−) mice

model, chondro/osteogenic markers are not up-regulated in the arteries prior to the

initiation of calcification[222]. One recent study found that calcium phosphate deposition

was a passive phenomenon and it was responsible for the osteogenic changes for the

VSMCs[223]. All these studies indicate that initial vascular calcification may be formed

without osteoblastic differentiation of VSMCs.

Page 98: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

80

Our study was designed to investigate the effect of calcified matrix on RASMCs’

phenotypic change. We hypothesize that the initial mineral deposition on elastin precedes

the osteoblast-like differentiation which is a pathological response to elastin degradation

and early arterial calcification. Our results showed that RASMCs lose their smooth

muscle lineage markers like SMA and MHC (Figure 4.2) and undergo

chondrogenic/osteogenic transformation (Figure 4.3) under calcified conditions. It is

unclear as to how calcified matrix triggers this response in RASMCs and further study is

needed to test this. Calcium and phosphorus levels in the media were similar in controls

and calcified matrix groups (data not shown), thus osteoblast-like transformation seen on

our studies was not due to higher amounts of free ions as shown by others[71, 208, 215].

We speculate that RASMCs synthesize small quantities of bone proteins (they come from

same mesenchymal origin of osteoblasts). These proteins in healthy state do not

accumulate in the vessel media; however, calcified elastin matrix can bind to these

synthesized proteins[224, 225] and increase local concentration causing RASMCs to turn

to osteoblast-like cells. Interestingly, our work, for the first time shows that upon removal

of calcified conditions, cells have the ability to revert back to SMC-like cell behavior.

When calcified matrix was removed from culture conditions, SMA and MHC expression

increased to normal levels, whereas osteogenic/chondrogenic markers decreased to

normal levels (Figure 4.4, Figure 4.5). This is important as we can then envision

demineralizing strategies to remove mineral from arteries and bring homeostasis to the

arterial structures.

Page 99: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

81

To summarize, there are possibilities of two distinctly different, yet

interconnected mechanisms to demonstrate the cause-and-effect relationship between

chondro/osteoblast-like differentiation of VSMCs and medial elastin-specific

calcification as shown in Figure 4.6. Model A: chondro/osteoblast-like differentiation of

VSMCs precedes medial elastin-specific calcification. Many factors could induce

VSMCs into osteoblast-like cells, such as elevated level of phosphate, lipids,

inflammatory cytokines and others[226]. Those differentiated osteoblast-like VSMCs

synthesize bone proteins such as osteocalcin, alkaline phosphatase to initiate mineral

deposition. This may be true in patients with chronic kidney disease and type-2 diabetes

where increase incidences of vascular medial calcification has been found[71, 219].

Model B: Medial elastin-specific calcification precedes chondro/osteogenic

differentiation of VSMCs. In ageing patients or in patients with diseases like aortic

aneurysm and arteriosclerosis, increased matrix metalloproteiases activity can lead to

accelerated elastic fiber degradation. This degradation would expose calcium binding

sites in elastin and allow initial mineral deposition[220]. This early mineral deposits may

anchor calcium binding proteins such as osteocalcin. This local increase in bone proteins

may lead to transformation of SMCs to osteoblast-like cells. It is also possible that these

two processes occur simultaneously and add mutually. Our current data mostly supported

Model B, that is, the first calcium deposition on elastin appear prior to the osteoblast-like

differentiation of RASMCs. Interestingly based on our data of reversal of osteogenic

markers, we hypothesize that demineralization of calcified arteries will return

homeostasis in arteries. In fact, recently we have shown that local delivery of chelating

Page 100: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

82

agent such as ethylene diamine tetraacetic acid (EDTA) led to regression of arterial

calcification in rats[227].

Figure 4.6. Model of putative links between medial elastin-specific calcification and

chondro/osteoblast-like differentiation of VSMCs in the artery. A: chondro/osteoblast-

like differentiation of VSMCs precedes medial elastin-specific calcification. Elevated

levels of phosphate, lipids, inflammatory cytokines and many other factors could induce

chondro/osteoblast-like differentiation of VSMCs. B: Medial elastin-specific calcification

precedes chondro/osteoblast-like differentiation of VSMCs. The elastin degradation

happens due to inflammatory conditions and matrix metalloproteinases. This in turn

exposes calcium binding sites on elastin causing first calcific deposits. This early

calcification leads to osteoblast-like differentiation in VSMCs that can further augment

calcification process.

Limitations of Current Study

There are some limitations to our studies. First, we used reagent grade synthetic

hydroxyapatite to mimic the ectopic deposition of mineral in vascular calcification, which

might not be ideal to represent the real pathologic condition of vascular calcification,

which shows poorly crystalline hydroxyapatite crystals and other calcium phosphate

minerals. We also used porcine aortic elastin that was allowed to calcify in vitro. It

Page 101: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

83

indeed showed poorly crystalline hydroxyapatite deposits along the elastin fibers. Finally,

we did the studies only for two time points: day 1, and day 7. It is still unknown if longer

exposure of RASMCs to calcified conditions would prevent reversal to normal

phenotype. More physiologic or animal studies are needed to further test the hypothesis

that medial elastin-specific calcification occurs first followed by cellular changes to

osteoblast like cells to augment mineral deposits.

4.5 Conclusion

In conclusion, our results demonstrate that hydroxyapatite and calcified elastin

induce chondrogenic and osteoblast-like differentiation of rat aortic smooth muscle cells,

and these studies support that elastin degradation and calcification may occur prior to

osteoblast-like transformation of VSMCs.

Page 102: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

84

Supplemental Figures

Figure 4.7. Experimental design.

Page 103: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

85

Figure 4.8. Graphical Abstract. At Day 0 rat aortic smooth muscle cells (RASMCs) were

seeded on petri dishes which have different substrate: two control groups, one was

normal media without substrate for hydroxyapatite (HA) study group, and another one

was pure elastin (PE) substrate for calcified elastin (CE). By Day 7 we found RASMCS

seeded on calcified matrix (Both HA and CE groups) had osteoblastic differentiation; at

Day 7 upon removal of calcified matrix, which was done by trypsinizing RASMCs first

and then re-seeding RASMCs on new petri dishes without any substrate but only normal

media, we found that differentiated RASMCs restored original phenotype by day 14.

Page 104: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

86

CHAPTER 5: EFFICACY OF CHELATING AGENTS TO REVERSE ELASTIN

SPECIFIC MEDIAL ARTERIAL CALCIFICATION

(This work has been published in

Calcified Tissue International 93, no. 5 (2013): 426-435.)

5.1 Introduction

Pathological calcification is defined as ectopic deposition of poorly crystalline

hydroxyapatite in soft tissues as observed in blood vessels and cardiac valves [228], The

two areas of calcification seen in arterial tissues include intimal calcification, detected in

atherosclerotic plaque present in the intima, and medial artery calcification of elastic

layers (MAC), detected in patients with diabetes, renal failure, and old age [6]; which is

independent of atherosclerosis. Medial calcification, termed as Monckeberg’s sclerosis,

mostly occurs along the elastic fibers of the media. Its presence correlates well with risk

of cardiovascular events and leg amputation in diabetic patients [229]. Currently no

clinical therapy is available to prevent or reverse this type of vascular calcification. Some

possible targets to block and regress calcification include local and circulating inhibitors

of calcification as well as factors that may ameliorate vascular smooth muscle cell

apoptosis [6]. Many of these approaches were focused on atherosclerotic calcification.

Almost no research has targeted reversing of elastin specific medial calcification[6].

Chelation therapy most often involves the injection of disodium ethylene diamine

tetraacetic acid (EDTA), a chemical that binds, or chelate ionic calcium, trace elements

and other divalent cations [141]. Chelation therapy has been touted as an effective

Page 105: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

87

treatment to reverse vascular calcification; however, it has been controversial with

opposing views [8-10]. At present no systemic scientific studies prove that it could

reverse cardiovascular calcification [9]. The Trial to Assess Chelation Therapy (TACT)

funded by the National Center for Complementary and Alternative Medicine began in

2003 to determine the safety and effectiveness of EDTA chelation therapy in individuals

with coronary artery disease has been finished by 2013. First published results showed

that intravenous chelation regimen with EDTA reduced the risk of adverse cardiovascular

outcomes but data was not sufficiently convincing to support the routine use of chelation

therapy for treatment of patients who have had an myocardial infarction [230].

Unfortunately, the efficacy of various chelating agents in reversing elastin specific

calcification from the peripheral vascular tissue is not studied very well either in vitro or

in animal experiments. Here we tested the hypothesis that common chelating agents, such

as disodium ethylene diamine tetraacetic acid (EDTA), diethylene triamine pentaacetic

acid (DTPA) and sodium thiosulfate (STS) can remove calcium from hydroxyapatite

(HA) and calcified tissues without damaging the tissue architecture.

5.2 Materials and methods

Chemicals

Hydroxyapatite, disodium ethylene diamine tetraacetic acid (EDTA), diethylene

triamine pentaacetic acid (DTPA) and sodium thiosulfate (STS), dichloromethane, were

all reagent grades and were purchased from Sigma-Aldrich Chemicals (St. Louis, MO).

Polyvinyl alcohol or PVA (Molecular Weight, M.W. = 15kDa) was purchased from MP

Page 106: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

88

Biomedicals Inc. (Solon, OH). Poly (L-lactic-co-glycolic acid) or PLGA

(lactide:glycolide = 50:50, M.W.= 60~80kDa) was a gift from Ortec, Inc. (Piedmont,

SC). Stock solutions of EDTA, DTPA and STS with concentration of 1, 5, 10 mg/mL

were prepared and stored at room temperature. Five mg/mL EDTA had pH of 4.8 while

5 mg/mL DTPA in 0.1 M NaOH had a pH of 12.2.

Tissues Preparation

In order to study chelation effect on elastin specific calcification, we wanted to

avoid effect of cells and other extracellular matrix components. Thus, we obtained pure

aortic elastin from porcine aorta with standard methods of purification[231]. We used

porcine aorta because of easy access and large quantities of elastin could be obtained.

Elastin homology is maintained in all species. In previous study, we showed that when

pure aortic elastin is implanted subcutaneously in rats, it undergoes calcification[200].

Thus, this calcified elastin could be used to study demineralization of elastin specific

calcification by chelating agents.

Calcified porcine aortic elastin (20 days explanted sample, 160~180 µg Ca/mg

tissue) was prepared by subdermal implantation of pure porcine aortic elastin in rats as

described previously [200]. Briefly, in juvenile male Sprague-Dawley rats (21 days old,

35 to 40 g; Harlan, Indianapolis, IN) a small incision was made on the back, and two

subdermal pouches were formed by blunt dissection. Each rat received two 30- to 40-mg

purified elastin implants (one per pouch), which were rehydrated in sterile saline 1 to 2

hours before implantation. The rats were sacrificed by CO2 asphyxiation after 20 days

Page 107: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

89

implantation and the implants were retrieved and stored at -80°C. Calcified human aorta

(100~300 µg Ca/mg tissue) was from cadavers (age 49-75, both male and female with

moderate or severe atherosclerosis) received from Greenville Hospital System

(Greenville, SC) following standard dissection technique. All samples were from the

abdominal aorta, just below the renal arteries.

Demineralization of Hydroxyapatite

To study optimal concentration chelating agents for best chelating efficacy, three

serial concentrations (1, 5 and 10 mg/ml) of chelating agents (STS, EDTA and DTPA)

were prepared. 10 mg of hydroxyapatite (HA) each was soaked in 1 ml chelating

solutions while the control was soaked in distilled and deionized water (DD water). For

the kinetic study of demineralization, Five mg of hydroxyapatite powder was suspended

in 30 mL solution of chelating agents (STS, EDTA, DTPA respectively, 5 mg/mL, n=3),

while the control group included DD water. At frequent intervals (2, 4, 6, 8, 24 hours), 1

mL of supernatant was withdrawn for Ca assay. Calcium content was measured by

atomic absorption spectroscopy (Perkin-Elmer Model 3030, Norwalk, CT) as described

previously [232]. Briefly, each sample (15–40 mg) was hydrolyzed in 1 ml of 6 N HCl in

a test tube at 70°C overnight and then the solution was completely evaporated at 70°C

with a continuous flow of nitrogen in the tube. The residue in the test tube was dissolved

in 1 ml of 0.01 N HCl. The hydrolyzed sample were diluted and analyzed by atomic

absorption spectroscopy with calcium standards.

Demineralization of Calcified Tissues

Page 108: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

90

Calcified porcine aortic elastin and separately calcified human aorta (10 mg each)

were treated with 30 mL chelating solutions (5 mg/mL of STS, EDTA, and DTPA, n=3).

The control group was treated with DD water both for calcified porcine aortic elastin and

calcified human aorta. At certain intervals (2, 4, 6, 8, 24 hours for porcine elastin and 8,

12, 24, 36, 48 hours for human aorta), 1 mL of solution was withdrawn for calcium assay.

Calcium content was measured by atomic absorption spectroscopy (Perkin-Elmer Model

3030, Norwalk, CT).

Alizarin Red Stain for Calcium

Calcified elastin samples were treated with chelating agents as described

previously and were then embedded in paraffin blocks, sectioned, and stained for

qualitative analysis of calcium with Dahl’s alizarin red stain. Since the calcified porcine

aortic elastin sample had more even distribution of calcification, we did the chelation

treatment to the bulk calcified elastin first and then made sections and stained the calcium

by Dahl’s alizarin red stain. For human aorta sample, since the distribution of

calcification was non uniform, we first sectioned samples and located calcification site

and then did the chelation treatment as described. Calcified human aorta were fixed in

10% neutral buffered formalin at room temperature for 24 h, processed with an automatic

tissue processor (Tissue TEK VIP, Miles Inc., Mishawaka, IN) and 6 µm paraffin

sections (n=12 sections per sample) were soaked in chelating solutions (5 mg/mL of STS,

EDTA, and DTPA, n=3) respectively for 4 hours. The control group was treated with DD

water (n=3). And then the slides were stained for 3 minutes at room temperature with 1%

Page 109: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

91

Alizarin red solution, and rinsed with distilled water. Sections were counterstained with

1% light green solution for 10 seconds and rinsed with distilled water and mounted.

Preparation of EDTA-loaded PLGA nanoparticles

EDTA-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles were prepared

by double emulsion method [233]. PLGA (200 mg) was dissolved in 2 mL

dichloromethane. EDTA (100 mg) was dissolved in 1 mL deionized water. For the

control group, no EDTA was used. Water-in-oil (W1/O) emulsion was prepared with an

Omni Ruptor 4000 ultrasonic homogenizer by 20 watts for 5 minutes. This prepared

primary emulsion was subsequently added to 10 mL of 1 % (w/v) PVA solution and the

second emulsion was prepared with homogenizer (20 watts, 5 minutes) to obtain water-

in-oil-in-water (W1/O/W2) emulsion. The nanoparticles were isolated by solvent

evaporation with stirring overnight at room temperature. After threes wash of deionized

water, the nanoparticles were lyophilized and stored at 4 ℃.

Measurement of loading efficiency and release profile of EDTA in PLGA

nanoparticles

5 mg EDTA-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles were

dissolved in 1 mL of dichloromethane and 5 mL distilled water was added and solutions

were vortexed for 30 minutes. After centrifugation at 13,000 rpm for 5 minutes the

supernatant were collected and the concentration of EDTA was spectrophotometrically

measured at 257 nm [234]. The loading efficiency is calculated by equation: EDTA

Page 110: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

92

loading efficiency = The amout of EDTA in nanoparticles/weight of nanoparticle *100%.

Five (5) mg EDTA-loaded PLGA nanoparticles were enclosed in dialysis units (Slide-A-

Lyzer® MINI Dialysis Units. Thermo Scientific, Rockford, IL) and incubated in 30 ml

PBS at 37 ℃ with mild agitation. At determined time intervals 1 ml sample was

withdrawn for quantification of EDTA release.

Local EDTA treatment in CaCl2 injury model of vascular calcification

Calcium chloride (CaCl2) injury model was used to create local arterial

calcification (abdominal aortic region) in rats [235]. 10 male Sprague-Dawley (SD) rats

(5-6 weeks old) were placed under general anesthesia (2% to 3% isoflurane), and the

infrarenal abdominal aorta was exposed and treated periadventitially with 0.20 mol/L

CaCl2 by placing CaCl2-soaked sterile cotton gauze on the aorta for 15 minutes. The area

was flushed with warm saline and wound site was closed with sutures. Animals were

allowed to recover and placed on normal diet. One week after first surgery, the abdominal

aorta was re-exposed in order to provide a treatment directly to the aorta. EDTA loaded

poly (lactic-co-glycolic acid) (PLGA) nanoparticle pellet (30 mg, with 5% EDTA

loading, n=5), and pure PLGA particle pellet (No EDTA entrapped, n=5) were placed

periadventitially. The rats were allowed to recover. After 1 week, rats were euthanized

and tissues (aorta) and blood were harvested and frozen in liquid nitrogen for plasma

calcium and phosphorus quantification. Calcium and Phosphorus level were measured by

atomic absorption spectroscopy as described previously [232].

Statistical Analysis

Page 111: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

93

Results are expressed as means ± SEM (standard error of the mean). Statistical

analyses of the data were performed using single-factor analysis of variance. Differences

between means were determined using the least significant difference with an α value of

0.05. Asterisks in Figures denote statistical significance (P < 0.05).

5.3 Results

Chelation of calcium from hydroxyapatite (HA)

Three series of concentration (1, 5, 10 mg/ml) of chelating agent solutions (STS,

EDTA and DTPA) were prepared in order to optimize the concentration needed for

efficient demineralization. HA powders (10 mg in each group) were suspended with

aqueous chelating agent solutions and the percentage HA solubilization was calculated

(Figure 5.1A). EDTA & DTPA were stronger chelating agent than STS, and could

remove more calcium from HA (Figure 5.1A). The chelating capacity was dependent on

concentrations of chelating agents with 10 mg/mL showing highest activity. 1 mg STS or

EDTA or DTPA chelate 1.3 mg HA, that is, 1 mole of STS, EDTA, DTPA chelate 0.41,

0.88, 1.02 mole of HA respectively.

Page 112: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

94

Figure 5.1. Demineralization of Hydroxyapatite. A: Effect of concentration of chelating

agents on dissolving calcium from hydroxyapatite powder. B: Kinetics of

demineralization of hydroxyapatite. All data are presented as means ±SEM, n=3 per

group.

Page 113: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

95

The concentration of 5 mg/mL of chelating agents were used for further studies

[236]. The kinetics of demineralization of hydroxyapatite is shown in Figure 5.1B. Most

of the chelation and removal of calcium occurred in first 2 hours and continued slowly up

to 24 hours for EDTA and DTPA. The chelating ability was: EDTA>DTPA>STS. Since

EDTA and DTPA solution as prepared had pH 4.8 and pH 12.2 respectively, we

examined the effect of pH on the demineralization of HA. HA was suspended in pH

buffers (pH 4.8 buffer: 59.0 mL 0.2M Sodium acetate and 41.0 mL 0.2M acetic acid; pH

12.2 buffer: 50 mL 0.2 M KCl and 20.4mL 0.2 M NaOH ). Calcium was not released

from HA in the pH range of 4.8 to 12.2 thus suggesting pH did not play a role in calcium

chelation. We also neutralized pH to 7.4 for all chelating solutions and still found similar

results of chelation (data not shown).

Chelation of calcium from calcified elastin

Calcified aortic elastin (3~5 mg, implanted subdermally in rats to calcify and then

isolated) were immersed in 1 mL solution of STS, EDTA, and DTPA with concentrations

of 5 and 1 mg/mL for 24 hours under vigorous shaking while the control group was

immersed in DD water (n=3 each group). Remnant calcium levels in the treated elastin

fiber were measured by atomic absorption spectroscopy. All chelating agents (STS,

EDTA and DTPA) could remove calcium from the calcified porcine elastin, with the

chelating ability sequence: EDTA ≥DTPA>STS (Figure 5.2A). This is consistent to the

previous HA results. For the EDTA treatment we calculated the percentage of calcium

loss by using the equation: the percentage of calcium loss = (the total calcium in calcified

Page 114: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

96

elastin - the remnant calcium in calcified elastin)/ the total calcium in calcified

elastin*100. Treatment with 1 mg/mL chelating agent for EDTA resulted in about 53 %

removal of calcium (Figure 5.2A), while higher EDTA concentration (5 mg/mL) was

effective in removing 75% of calcium (Figure 5.2A). Such increase in calcium

dissolution was not found in DTPA and STS groups after increasing chelating agent

concentrations (Figure 5.2A).

The kinetics of demineralization of calcified elastin for chelating agents with the

concentration of 5 mg/mL is shown in Figure 5.2B (n=3 in each group). Both EDTA and

DTPA were effective in removal of calcium from calcified porcine elastin while STS was

not effective, which corroborated with the result of hydroxyapatite. However; kinetics of

calcium removal from calcified elastin was much slower than HA. Longer incubation

times lead to continuous removal of calcium so that within 24 hours EDTA could remove

almost all calcium from calcified elastin (Figure 5.2B). Dahl’s alizarin red stain at 24

hours of treatment further confirmed that EDTA treatment could remove all calcium

while DTPA was partially successful and STS treatment was ineffective (Figure 5.2C).

Page 115: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

97

Figure 5.2. Demineralization of calcified elastin. A: Optimal concentration of chelating

agents needed to remove Ca from calcified elastin. B: Kinetics of demineralization of

calcified porcine elastin. All data are presented as means ±SEM, n=3 per group. c: Dahl’s

alizarin red stain for porcine elastin after chelation treatments. Alizarin red stains calcium

red. Only EDTA treatment removed calcium from calcified elastin.

Page 116: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

98

Chelation of calcium from calcified human aorta

The kinetics of extraction of calcium from calcified human aorta after treating

with chelating agents for 48 hours, showed that EDTA and DTPA were most effective in

removing calcium while STS was not effectively similar to what was observed in HA and

elastin studies presented above (Figure 5.3A). The kinetics of calcium dissolution was

slower probably due to denser nature of aortic tissue. To study tissue architecture after

chelation, 6 µm sections of human aorta were treated with chelating solutions for 4 hours.

Dahl’s alizarin red stain (Figure 5.3B) for the human aorta sections showed that EDTA

and DTPA removed the calcium while STS treatment was not effective. The tissue

sections maintained their typical arterial integral structure. Chelation treatment did not

cause delamination or removal of certain parts of artery for all chelating agents treated

group (STS, EDTA, and DTPA) comparing to the control group.

Page 117: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

99

Figure 5.3. Demineralization of calcified human aorta. A:Kinetics of demineralizaion of

calcified human aorta. All data are presented as means ±SEM, n=3 per group. B: Dahl’s

alizarin red stain for human aorta sections after treatment with chelating agents showing

efficacy of EDTA and DTPA in removal of calcium.

Page 118: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

100

Reversal of arterial calcification in vivo by EDTA

As EDTA was found to be most successful in removing calcium in vitro, we

tested its efficacy in removing calcium from calcified aorta in vivo. We prepared PLGA

nanoparticles loaded with EDTA so that prolonged (24 hour) controlled release of EDTA

could be achieved when the nanoparticles are placed close to the calcified aorta. Our in

vitro release profile showed that EDTA could be released from PLGA nanoparticles

within 3 days (Figure 5.4). Next, we tested if local delivery of EDTA could remove

calcium from calcified rat aorta in vivo. We have shown previously that when abdominal

aorta of rats were exposed to chemical injury with calcium chloride, it lead to elastin

specific medial arterial calcification with time [199]. We used 7 day time point after

injury for EDTA therapy as substantial calcification is seen in this model at that time

[15]. After 7 days, EDTA loaded PLGA nanoparticle pellets (30 mg with 5% EDTA

loading, thus dosage was 0.01 g EDTA/kg) were placed on the aorta. We observed that

the PLGA particles were spontaneously absorbed onto the wet abdominal aorta. Seven

days after initial EDTA treatment rats were sacrificed to test if local EDTA therapy

would reverse elastin calcification. In vivo studies were continued for 7 days to make sure

all of the EDTA could be released and available to remove calcium. Both calcium and

phosphorous level were significantly lower in the EDTA treated group as compared the

control group with blank nanoparticles (Figure 5.5A). Histology analysis also showed

that the aortic calcification was greatly reversed in the EDTA treated group (Figure

5.5B). The calcium and phosphorus level of the blood plasma in both groups were also

quantified and there was no statistical significant difference between the EDTA treated

Page 119: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

101

group and pure PLGA control group (Figure 5.5C). Clearly suggesting that local delivery

of EDTA did not change serum calcium levels, which is often the case with systemic

delivery.

Figure 5.4. EDTA release profile from PLGA nanoparticles. All data are

presented as means ±SEM, n=3 per group.

Page 120: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

102

Figure 5.5. Reversal of arterial calcification in vivo by EDTA. A: Calcium and

phosphorus level of the harvested abdominal aorta after treatment of PLGA particles.

Study group: EDTA-loaded PLGA. Control group: PLGA alone. Asterisks in Figures

denote statistical significance (P < 0.05) comparing to control group without EDTA

entrapped. B: Alizarin Red stains for the harvested aorta treated by EDTA-loaded PLGA

particles and control PLGA particles. C: Calcium and phosphorus level of the rats’

plasma after treatment of PLGA particles. There is no statistical significant difference

(P>0.05) comparing to control group without EDTA entrapped. All data are presented as

means ±SEM, n=5

Page 121: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

103

5.4 Discussion

This is the first study to compare three types of chelating agents (STS, EDTA, and

DTPA) used for chelation therapy and their chelating efficiency to remove calcium from

the hydroxyapatite, calcified aortic porcine elastin and calcified human aorta. We found

an optimal concentration (1~10 mg/mL) of chelating agents works very well to remove

calcium. Our results indicate that both EDTA and DTPA could effectively remove

calcium from hydroxyapatite and calcified tissues, while STS was not effective. The

tissue architecture was not altered during chelation. Furthermore, our in vivo study

demonstrates the proof of the concept for the first time that local delivery of chelating

agents such as EDTA could be used to reverse medial aortic calcification which is found

in arteriosclerosis.

Chelating agents’ effectiveness in removing plaques has been advertised and used

in certain countries, however, the treatment is not FDA approved in US because lack of

substantial conclusive data. The actual published data is very scarce in both animals and

human patients and no comparative in vitro or animal studies have been performed to

demonstrate the calcium chelating ability of different types of chelating agents (STS,

DTPA and EDTA). Pasch et al [119] found that STS could probably chelate calcium by

forming soluble calcium thiosulfate complex. They found that it prevents vascular

calcification in uremic rats. But systemic treatment of STS also lowered the bone strength

in animals probably due to STS chelation of bone HA. They proved direct calcium

Page 122: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

104

chelation by STS through a decline of serum ionized calcium concentrations when

exposed to STS in vivo and in vitro. Adirekkiat et al[120] demonstrated that STS could

remove calcium from precipitated minerals and delayed the progression of coronary

artery calcification(CAC) in hemodialysis patients. Patients with a CAC score≥300 were

included to receive intravenous sodium thiosulfate infusion twice weekly post-

hemodialysis for 4 months. They found that CAC score did not change significantly in

the STS treatment but increased substantially in the control group. Our present study

shows that the chelating ability of EDTA and DTPA was superior to STS to remove

calcium from hydroxyapatite like mineral from calcified elastic tissue.

Disodium EDTA is an amino acid used to sequester divalent and trivalent metal

and mineral ions (e.g., lead, mercury, arsenic, aluminum, cobalt, calcium etc.). EDTA

binds to metals via 4 carboxylate and 2 amine groups [237]. Diethylene triamine

pentaacetic acid (DTPA) is consisting of a diethylenetriamine backbone with five

carboxymethyl groups. The molecule can be viewed as an expanded version of EDTA

and it is used similarly for metal chelation. Cavallotti et al [238] found that intravenous

injection of EDTA substantially reduced the extent of calcium deposits in the aortic wall.

The general decalcifying mechanism of EDTA may derive from its affinity for divalent

ions, especially calcium. Maniscalco et al [239] demonstrated that calcification in

coronary artery disease could be reversed by EDTA–tetracycline long-term therapy.

Their hypothesis was that calcification is caused by nanobacteria (NB). In that study,

EDTA was given as a rectal suppository base every evening, which was shown to result

in high blood EDTA levels sustained for a long time. They proposed that NB were

Page 123: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

105

sensitive in vitro to tetracycline and its action is increased by EDTA dissolving NB

apatitic protective coat. Thus, they concluded that combination of these drugs might offer

a novel treatment for calcific atherosclerotic disease. EDTA has also been proven to

reduce the risk of calcific tendinitis of the shoulder [237] if used as a chelating agent. In

that study EDTA was administered through single needle mesotherapy and 15 minutes of

pulsed-mode 1 MHz-ultrasound. Calcifications disappeared completely in 62.5% of the

patients in the study group and partially in 22.5%; calcifications partially disappeared in

only 15% of the patients in the control group, and none displayed a complete

disappearance.

Despite these clinical studies and other EDTA chelation therapies that have gone

to clinical trials since 2003[240], little basic research is done to study chelation of

hydroxyapatite like mineral in vitro and in animal studies. This might be one of the

reasons why lots of negative opinions directed toward the chelation therapy for removal

of calcium from atherosclerotic plaques. Dr. Jackson[8], the editor of International

Journal of Clinical Practice, stated that “Chelation therapy is TACT-less, one wonders

why it is still widely used, given the lack of evidence of benefit and potential to do harm

(financial gain seems the most likely reason)”. Atwood et al[9] concluded that “…TACT

is unethical, dangerous, pointless, and wasteful. It should be abandoned.” Fraker et al[10]

concluded that “Chelation therapy (intravenous infusions of ethylenediamine tetra-acetic

acid or EDTA) is not recommended for the treatment of chronic angina or atherosclerotic

cardiovascular disease and may be harmful because of its potential to cause

hypocalcaemia.” First randomized clinical trial results published this year for EDTA

Page 124: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

106

chelation therapy for coronary artery disease showed some improvements but the data

was not statistically significant to warrant use of chelation therapy for all patients with

myocardial infarction [8]. In all clinical studies, chelation therapy was tested to remove

calcium from atherosclerotic plaque and improve heart function. Atherosclerotic plaque

calcium deposits are due to necrotic tissue core and removal of calcium from plaque may

not remove cholesterol laden fatty deposits, thus may not be very effective in reducing

heart burden. Some have suggested that calcification make plaques more stable and

removal of calcium may make them vulnerable to rupture and that may have led to

negative connotations. Our present study with chelating agents is necessary and

significant because such therapy was never tested suitably to remove calcium from

medial arterial calcification that occurs in absence of atherosclerotic plaques in patients

with diabetes, end stage renal disease, and old age. In those cases calcification is

observed along the medial elastic layers that make arteries stiff. We envision using

chelation therapy for such type of calcification.

Our current results show that both EDTA and DTPA could effectively remove

calcium from calcified aortic elastin and calcified human aorta, while STS is not so

effective. We have demonstrated that vascular elastin specific calcification could be

reversed by chelating agents in vitro. Our in vivo study in the CaCl2 injury model, for the

first time, shows proof of the concept that chelating agent loaded PLGA nanoparticles

could reverse medial elastin-specific aortic calcification. One criticism of chelation

therapy is its effects on serum calcium and bone HA. One previous study of systemic

administration of STS in rats showed that such systemic chelation treatment led to the

Page 125: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

107

decrease of serum calcium, phosphate and also compromised bone integrity. STS

treatment group required significantly lower load to fracture the femurs as compared to

controls[236]. Another study in humans also showed that systemic administration of

chelation agent caused decrease in serum calcium and serum phosphate levels and caused

bone loss by increasing endogenous parathyroid hormone secretion[14]. We consider

chelation therapy could be useful in patients with diabetes and old age to remove elastin

specific calcification if chelating agents such as EDTA are protected in polymeric

capsules (such as nanoparticles) and delivered at the site or targeted to vascular

calcification. Such treatment would prevent serum calcium chelation and undesired effect

on bone density. Our result in rats study showed such local EDTA treatment with PLGA

nanoparticles, completely reversed calcification without damaging vascular structure.

This is for the first time anyone has shown effectiveness of local therapy in reversal of

calcification. Such local EDTA treatment also did not change serum calcium and

phosphorus levels; thus could be used without systemic effects. This is because local

delivery method used in this study had much lower dose of chelating agents, specifically

0.01 g EDTA/kg rats comparing to those systemic administration of 0.4 g STS/kg

rats[236] and 0.04 g EDTA/kg human[230].Our long term goal is to develop site-specific

delivery of the chelating agents with targeted nanoparticles to reverse vascular

calcification without causing the negative impact on blood chemistry and bone integrity.

Page 126: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

108

Limitations of Our Study

There are some limitations of our current study. Firstly, for in vivo chelation study

we did not measure plasma level of other biochemically important metal ions such as

chromium, zinc, cobalt, and copper and toxic metals, such as cadmium and lead, which

might be affected by the chelation treatment [241]. We used local delivery of small

concentrations of EDTA and thus systemic concentrations of chelating agent would be

negligible and may not affect other metal ions. Secondly, our animal study results might

not be directly applicable to patients with chronic kidney disease or patients on dialysis.

In our animal study, the rats had normal kidney function while those patients with

arteriosclerosis typically have non-regulated plasma level of calcium, phosphorus and

other ions[242]. Finally, we did not look at the bone integrity which might affected by the

EDTA treatment. Again, because we used local therapy of EDTA at very low

concentrations (4 fold lower than used in clinical trials, per kg basis) instead of systemic

delivery, we hypothesize that it will not affect bone integrity.

5.5 Conclusion

In conclusion, we did a comparative in vitro study for three types of chelating

agents (STS, EDTA and DTPA) and showed that EDTA and DTPA could effectively

remove calcium from calcified aortic elastin and calcified human aorta. We, for the first

time, show that local EDTA treatment with PLGA nanoparticles could reverse

calcification without causing vascular damage and change in the normal plasma level of

calcium and phosphorus.

Page 127: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

109

Supplemental Figures

Figure 5.6. Types of chlating agents and chemical structure.

Figure 5.7. Surgery protocol and photos.

Reversal of aortic calcification in vivo by EDTA

Page 128: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

110

CHAPTER 6: SYSTEMIC EDTA CHELATION THERAPY FAILED TO

REVERSE MAC AND HAD SIDE EFFECTS

6.1 Introduction

Vascular calcification is an independent risk factor for cardiovascular morbidity

and mortality. Vascular calcification occurs at two different sites- intima and media.

Intimal calcification is generally associated with atherosclerotic plaques and

inflammation and can occlude the lumen while medial calcification (MAC) occurs in the

elastin lamina of medium and large arteries in patients with chronic kidney disease (CKD)

or diabetes causing increase in arterial stiffness. Surgical methods like directional

atherectomy and stent grafts are used to treat atherosclerosis and intimal calcification and

to open occluded arteries [4, 5]. However, there are no treatments available for medial

elastin-specific calcification.

EDTA (disodium, ethylenediamine tetraacetic acid) chelation therapy has been

tried for reversing intimal vascular calcification with some success [17, 168, 172]. EDTA

binds or chelates ionic calcium (Ca), trace metals, and other divalent cations [141]. Many

possible mechanisms are proposed for EDTA chelation therapy to reverse calcification.

These mechanisms include: directly removing calcium deposits from calcified arteries

[134], binding serum calcium to reduce calcification, chelating heavy metals, and

normalizing Ca metabolism by reactivation of enzymes inhibited by toxic heavy metals

[135-138]. However, at present effectiveness of EDTA chelation therapy is still disputed

[9]. Most of the EDTA treatments were performed by subcutaneous infusion, or systemic

Page 129: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

111

application of EDTA solution at very high dose (around 40 ~ 50 mg/kg) [11-17]. Many

side effects have been found with such a high systemic dose of EDTA chelation therapy

including: renal toxicity [18-23] , hypocalcemia [14, 24, 25], and bone loss [14, 26];

however, side effects are variable.

No animal studies are performed as yet to test if systemic EDTA therapy can

reverse elastin specific medial calcification (MAC). We created vascular medial elastin

specific calcification by abdominal application of calcium chloride to the abdominal aorta

of rats and tested if systemic EDTA therapy can reverse this type of calcification [27, 28].

We also analyzed possible side effects to serum Ca, urine Ca, bone loss, and kidney

toxicity.

6.2 Materials and methods

CaCl2 injury model and EDTA administration

An illustration of the experimental design is shown in Figure 6.1. All procedures

and treatments in animal studies were approved by the Institutional Animal Care and Use

Committees (IACUC) at Clemson University.

Page 130: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

112

Figure 6.1. Experimental protocol for CaCl2 injury model and EDTA treatment of rats.

CaCl2 injury model (represent as symbol of arrow) was bone by a single periadventitial

0.5 M CaCl2 injury to abdominal aorta (15 minutes treatment) at starting time point of

experiment (Day 0, week 0). There were two treatment groups: 1) saline as control group

(n=5, 5 rats), tail vein injections with saline 2 times a week for another 2 weeks. 2)

EDTA as study group (n=5, 5 rats), tail vein injections with EDTA (50 mg/kg) 2 times a

week for another 2 weeks. Rats were placed in metabolic cages for urine collection for 24

hours (Day 21); after that all rats were given another injection (Day 22) of saline or

EDTA (depending on individual treatment groups) and then urine samples were taken at

time points of 1, 2, 3 and 4 hours post injection; finally rats were euthanized and organs

were harvested (represent as symbol of six-point stars).

CaCl2 injury model was used to mimic local aortic calcification at the abdominal

aortic region following previous protocol [27, 28]. Briefly, ten male Sprague-Dawley (SD)

rats (6 to 8 weeks old) were anesthetized by 2-3% isoflurane. A ventral laparotomy

exposed the peritoneal cavity. A 15 mm section of the infrarenal abdominal aorta was

exposed and injury was created by placing 0.5 M CaCl2 soaked sterile cotton gauze on

top of the aorta for 15 minutes. After CaCl2 application, the gauze was removed and the

area was briefly flushed with warm saline. Incisions were then closed with continuous

Page 131: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

113

sutures (Abdominal and subcutaneous) and surgical staples. Then rats were allowed to

recover and maintained in standard chow conditions for one week.

One week after CaCl2 injury, rats were randomly separated into two groups: 1)

control saline group and 2) EDTA group (n=5/group). Either saline or EDTA (50 mg/kg

EDTA in saline) was delivered intravenously 2 times a week, for 2 weeks by tail vein

injections The dosage for EDTA was chosen based on other animal studies and clinical

trials for EDTA therapy [11-17].

On the final day of treatments (Day 21), all rats were placed in metabolic cages

(fasting for a day - removal of food only, water supply not removed) for urine collection.

The day immediately following fasting (Day 22), all rats were given another injection of

saline or EDTA (depending on individual treatment groups) and then urine samples were

taken at time points of 1, 2, 3, and 4 hours post injection and kept in – 80 ℃ for further

analysis. Immediately after urine collection all rats were euthanized by CO2 asphyxiation

and all organs including the aorta (from the heart to the iliac bifurcation), blood, femoral

bone, kidney, heart, liver and spleen were harvested. Heparin (10 units/ml blood) was

used as anti-coagulant for blood samples and serum was isolated and stored at – 80 ℃ for

further assay. Aorta, kidney, heart, liver and spleen were fixed in 10% buffered formalin

for further analysis.

Evaluation of aortic calcification

Aortic calcifications were evaluated by stereomicroscope micro-computed

tomography (micro-CT) scanner, histology, and atomic absorption spectrometry (AAS).

Page 132: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

114

Aortas from all 10 rats were cleaned of all adherent tissues by blunt forceps after

formalin fixation. Plastic tube (PE, 1 mm outside diameter) was inserted into the cleaned

aortas for easier handling during imaging.

Whole aortas were imaged by stereomicroscope (Leica M80 stereo microscope,

Leica Microsystems Inc. Buffalo Grove, IL) both before and after alizarin red staining

[96, 243, 244]. Briefly, whole aortas were soaked in freshly made 2% alizarin red

solution (pH 4.1-4.3, pH) for 10 minutes and washed by distilled and deionized water for

another 5 minutes. Then aortas were then imaged by stereomicroscope.

Micro-CT imaging for harvested aortas was based on modified protocol [237,

239]. Micro-CT was done by using Siemens Inveon microPET/CT scanner (Preclinical

Solutions, Siemens Healthcare Molecular Imaging, USA). Briefly, a 360-degree axial

scan was collected at a peak tube potential of 80kVp and tube current of 500uA. Data

were reconstructed to 100 μm pixel size using a Feldkamp cone beam algorithm. Virtual

2D cross-sections and 3D maximum intensity projection views were created using

MeVisLab software (MeVisLab, Mevis, Bremen, Germany). Exported movies from

MeVisLab were further edited by video-editing software- Corel VideoStudio Pro

X7(Corel Inc, Mountain View, CA). (Available as online video supplement 1).

Following the non-invasive imaging by stereomicroscope and micro-CT scanner

mentioned above, aortas were processed for histology with alizarin red staining for

visualization of calcium. Briefly, aortas were fixed in 10% buffered formalin, processed

with an automatic tissue processor (Tissue TEK VIP; Miles, Mishawaka, IN) and then

Page 133: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

115

sectioned into 6 μm paraffin sections for Alizarin red staining. Slides were stained in

freshly made 2% alizarin red solution (pH 4.1-4.3) for 3 minutes, washed by distilled and

deionized water and then counterstained with 1% light green solution for 10 sec and

mounted for optical microscope examination.

Remaining parts of abdominal aortas were used for Ca quantification by AAS. [28,

232]. Briefly, 2 ~ 10 mg of abdominal aortas were hydrolyzed in 1mL of 6N Ultrex HCl

(Baker Company, Phillipsburg, NJ) and then re-suspended in 0.01N Ultrex HCl.

Hydrolyzed samples were diluted and analyzed with AAS for calcium.

Calcium in serum and urine

Serum and urine samples were diluted before Ca quantification by AAS. Same

protocols were used as mentioned above for AAS.

Bone Analysis by Micro-CT and three points bending

The rat femoral bones were dissected and fixed in 10% buffered formalin from

both groups (saline group and EDTA group, n=5 each). The integrity of femoral bone

was evaluated by Micro-CT and by functional stiffness and fracture studies. Stiffness and

maximum load were tested based on three points bending mechanical test. Micro-CT for

rat femoral bones was performed by the same protocol as aortas mentioned earlier.

Representative scanning data for the whole 2D view slices from each group (saline group

and EDTA group) is available as an online supplement video 2.

Page 134: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

116

Three points bending test was performed using Bose test machine (Electroforce

3200, Bose, MN, USA) equipped with a 450N load cell. Rat femoral bones were placed

in the machine (available as online supplement video 3 to show the fixture and fracture

locations). The stiffness was tested by measuring bone displacement (in mm) at a given

force (N). Later, bones were exposed to higher force until fracture occurred from which

the maximum enduring load (N) was determined.

Statistical analysis

Results are expressed as means ± standard deviation (SD). Statistical analyses of

the data were conducted using single-factor analysis of variance. Differences between

means were determined using the least significant difference with an α value of 0.05.

Asterisks in Figures represent statistical significance (P < 0.05).

6.3 Results

Systemic EDTA chelation therapy did not reverse aortic calcification

Periarterial application of calcium chloride led to medial elastin specific

calcification in the abdominal aorta. Whole rat aorta when stained with alizarin red

showed intense red staining for calcification where injury was created. Histology of

cross-sections of aorta confirmed elastin specific medial calcification (Figure 6.2A). Two

weeks of systemic EDTA chelation therapy did not reverse aortic calcification (Figure

6.2) as assessed by stereomicroscopy of whole aorta, histology, and calcium

quantification methods (AAS). Results showed no visible difference in calcification level

between saline study group and EDTA study group (Figure 6.2A). Ca quantification in

Page 135: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

117

aortas by AAS also did not show statistical differences (P = 0.6491, n = 5) between

EDTA treated group and saline group confirming that 2 weeks of EDTA treatment was

ineffective in removal of aortic calcium (Figure 6.2B).

Page 136: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

118

Figure 6.2. Evaluation of aortic calcifications by stereomicroscope, histology and AAS.A:

Photos for aortas took by stereomicroscope both before and after staining with alizarin

red and also one representative images of histological section after alizarin red staining

(n=5). B: Ca quantification in aortas by AAS. Micro-CT data were available as online

video supplementary materials.

Micro-CT results for aorta corroborated histology and no visible differences

between saline study group and EDTA study group were found (online video supplement

1).

Systemic EDTA chelation therapy led to hypocalcemia and hypercalciuria

Systemic EDTA chelation therapy caused side effects that influenced the Ca

amounts in serum and urine (Figure 6.3). Ca amount in the rats’ serum for EDTA study

group were significantly lower than the saline control group (Figure 6.3A, P = 0.0045, n

= 5). On the contrary, Ca amount in rats’ urine at the end (Day 22) after two weeks

EDTA treatment were significantly higher than saline control group (Figure 6.3B, P =

0.0001, n = 5). After EDTA application, urinary total Ca excretion increased

continuously for 4 hours (Figure 6.3C).

Page 137: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

119

Figure 6.3. Ca quantification in serum and urine by AAS. A: Ca amounts in serum. B: Ca

amounts in urine at the end day (Day 22). C: Changes in urinary Ca concentrations post

saline and EDTA application.

Systemic EDTA chelation therapy did not cause bone loss

Since hypercalciuria (excessive urinary calcium excretion) induced by EDTA

application can potentially cause bone loss, we did morphology and functional test for

rats’ femoral bones both for EDTA treated and untreated groups. Micro-CT for rat

femoral bones selectively at 15 mm below the trochanter major showed no visible

difference, such as reduced mineralization or cystic transformation, between EDTA-

Page 138: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

120

treated and untreated groups (Figure 6.4A). 2D imaging of virtual-slices (Total about 500

slices, 100 μm resolution) of rats’ femoral bones also indicated no visible differences for

all virtual-slices (online supplement video 2). Functional studies on bone integrity by

three points bending test revealed no significant changes to stiffness (measured as bone

displacement at a given force, N/mm) of rat femoral bones for EDTA treated group

compared to untreated group (Figure 6.4B, P = 0.8309, n = 3). Interestingly, EDTA

treated groups even had slightly higher maximum load compared to untreated group

(Figure 6.4C, P = 0.0273, n = 3).

Page 139: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

121

Figure 6.4. Bone integrity of EDTA-treated (study group) and untreated rats. A: Rats’

femoral bone morphology by Micro-CT. No visible difference were seen such as reduced

mineralization or cystic transformation for EDTA-treated and untreated groups. B:

Stiffness of rats’ femoral bones analyzed from three point bending test. C: Maximum

(Max) load of rats’ femoral bones analyzed from three point bending test.

6.4 Discussion

The present study demonstrates that systemic EDTA chelation therapy does not

reverse local elastin-specific medial aortic calcification in rats. We further show that

systemic EDTA chelation therapy caused hypocalcemia (low serum calcium levels) and

hypercalciuria (excessive urinary calcium excretion) but did not cause bone loss. These

Page 140: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

122

results indicated that systemic EDTA chelation therapy might not have therapeutic effects

in aspects of reversal of elastin-specific medial calcification that is found in patients with

CKD and chronic diabetes.

One of the proposed mechanisms for EDTA chelation therapy to reverse

calcification suggests that EDTA could directly remove calcium deposits from calcified

arteries [134]. We recently have demonstrated that EDTA could effectively remove Ca

from hydroxyapatite and calcified elastin and human aorta tissue In vitro [28]. Animal

studies with EDTA treatment have shown mixed results. For example, Kaman et al and

Cavallotti et al. showed that systemic infusion of EDTA significantly reduces calcium in

aortas in atherosclerotic rabbit model [11, 13]. But O'Brien et al showed that EDTA

chelation therapy (40 mg/kg, daily intravenous injection, 4 weeks) showed a 74%

increase in triglyceride levels but no benefit on arterial lesions in an atherosclerotic,

obese, diabetic rat model [165].

Many clinical studies have investigated the therapeutic effects of EDTA chelation

therapy with mixed results [15, 16, 142, 167]. Most of them were focused on other

aspects such as improvement in clinical symptoms such as cardiac output but not on

specifically reversal of calcification. Olszewer et al. in 1990 [137] did a pilot double

blinded study with EDTA chelation therapy (1.5 g/ EDTA per patient, 20 intravenous

infusion) with 10 patients (all male, aged 41 to 53 years) diagnosed with peripheral

vascular disease. Results showed that EDTA chelation therapy had substantial

improvements for the walking test and master exercise test. In 2005, Shoskes et al. [170]

Page 141: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

123

reported a case study involving 16 male patients with recalcitrant chronic

prostatitis/chronic pelvic pain syndrome (CPPS) showing significant improvement of

symptoms of recalcitrant CPPS in the majority of the men after ComET therapy, which

was a combined EDTA chelation therapy with other drugs. One recent randomized trial

done by Lamas et al. in 2013 [17] with 1708 patients with myocardial infraction showed

that EDTA chelation therapy (50 mg/kg EDTA, intravenous infusion , one time a week,

40 weeks) would modestly reduce the risk of adverse cardiovascular outcomes but the

results did not lead to statistical significance. Further, this study did not highlight calcium

score data.

None of the previous studies, either in animals or in a clinical study, looked at

whether EDTA can remove calcium from medial elastin specific medial crterial

calcification (MAC). This condition can cause significant increase in arterial stiffness and

hypertension in patients. Thus, we tested EDTA chelation in an animal model where

elastin specific medical calcification is observed [27]. Local periadventitial treatment of

infrarenal aorta allowed us to induce elastin specific arterial calcification independent of

atherosclerosis similar to what is found in CKD patients. We chose EDTA doses similar

to dose used previously in other animal studies and in chelation therapy in patients [11-

17]. However, the two week treatment period of our study was shorter than others who

investigated 4 week [12] to 9 week [14] treatment periods. We chose shorter period due

to less severe calcification seen in our model at seven days. Our results indicated EDTA

chelation therapy by systemic infusion might not be effective to reverse elastin-specific

medial calcification possibly due to site of calcification, which is in the medial layer of

Page 142: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

124

the artery and may not accessible to systemically delivered EDTA under high blood flow

conditions in the aorta. The half-life of EDTA in human beings was 20 to 60 minutes and

that EDTA was excreted primarily by the kidneys with about 50% excreted in one hour

and over 95% with 24 hours [245]. Similar data for rats is not available at this time but

half-life could be even less than humans. Thus, intravenously delivered EDTA may not

be as effective.

Many side effects are associated with systemic EDTA chelation therapy due to

high dosage. The side effects include are but not limited to: hypocalcemia [14, 24, 25],

bone loss [14, 26] and renal toxicity [18-23]. Our current results in rats also showed side

effects of hypocalcemia (low serum calcium levels) and hypercalciuria (excessive urinary

calcium excretion) (Figure 6.3). However, our results did not show any bone loss either

by morphology using Micro-CT or functional test using three points bending mechanical

test (Figure 6.4). Rudolph et al. in 1988 [26] conducted a case study with 61 patients and

showed that EDTA chelation therapy (3 g EDTA, 20 intravenous infusion, 5-9 weeks)

had no effect on bone density and may even enhance bone growth in patients with

osteoporosis. One proposed mechanism for EDTA’s effect on bone integrity is that

EDTA chelation therapy will lower serum calcium, and this would stimulate the

parathyroid to release parathormone thus pulling Ca out of the bones. However, because

the release of parathormone is pulsatile, an increase, rather than decrease, of new bone

formation would occur [172]. Whether this theory was correct or not is still unclear.

However, our present results indicated that bone loss was not a major adverse event for

EDTA chelation therapy。 In fact we did find slight increase in maximum load for bone

Page 143: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

125

fracture in EDTA group suggesting increase in bone density. However, the major

possible side effects for EDTA chelation therapy were hypocalcemia, and hypercalciuria

in our study. However, we did not see EDTA treatment induced renal toxicity (Shown in

supplementary data, H&E staining of rats’ kidney) such as renal tubular vacuolization

and pinocytosis by the chelate as shown by others [18-23]. This might because our 2

weeks’ EDTA treatment is too short to induce renal toxicity.

Since systemic EDTA chelation therapy has so many possible side effects, it

would be important to further investigate how to avoid such side effects. One possible

way is using targeted EDTA chelation therapy. In fact, we previously showed that local

placement of EDTA loaded nanoparticles close to calcified aorta in calcium chloride

mediated injury model could reverse aortic calcification without causing side effects like

hypocalcemia [28]. Of course, such surgical approach is too invasive and would not be

feasible clinically. Our lab is currently working on targeted delivery of EDTA loaded

nanoparticles, which may provide a promising therapy to reverse elastin-specific arterial

calcification at much lower doses thus preventing systemic side effects.

Limitations of Current Study

There are some limitations to our studies. Firstly, our EDTA chelation therapy

was performed for a period of only two weeks. We do not know if long-term EDTA

treatment will have therapeutic effects to reverse vascular medial calcification. Secondly,

we did not investigate possible side effects besides serum calcium, urine calcium, and

bone loss. More kidney functional evaluation might be necessary such as serum

Page 144: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

126

creatinine level. Finally, we only evaluated EDTA chelation therapy in rat CaCl2 injury

model, which induces local medial calcification at the site of injury. Patients have

calcification dispersed throughout arterial tree thus calcification animal models such as

warfarin [96]or vitamin D [99] application where calcification is seen at various places

such as aortic arch and renal and femoral bifurcation might be necessary in order to

investigate EDTA chelation therapy’s therapeutic effects.

6.5 Conclusions

Our results demonstrated that systemic EDTA chelation therapy for 2 week period

did not reverse local elastin specific aortic calcification in rats. We also showed that such

therapy caused side effects of hypocalcemia and hypercalciuria but it did not cause bone

loss. Thus, reversal of medial elastin calcification may not be achieved by short-term

systemic EDTA therapy. As such calcification is seen patients with CKD who already

have diseased kidney, further toxicity caused by EDTA could be detrimental to patients’

health.

Page 145: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

127

Supplemental Figures

Figure 6.5. H&E staining for rats’ kidneys. No treatment induced histological change

was found.

Page 146: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

128

Figure 6.6. One representative aorta’s Micro CT images from two groups.

Page 147: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

129

Figure 6.7. Three points bending test: before and after fracture.

Page 148: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

130

Figure 6.8. Load – Displacement curve for three points bending.

Page 149: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

131

Figure 6.9. Micro-CT for rats’ femoral bone.

Page 150: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

132

Figure 6.10. Micro-CT for three slices of rats’ femoral bone.

Page 151: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

133

CHAPTER 7: TARGETED EDTA CHELATION THERAPY TO REVERSE

CALCIFICATION

7.1 Introduction

Medial arterial calcification (MAC), termed as Monckeberg’s sclerosis, is a type

of vascular calcification disease that mostly occurs as linear deposits along elastic

lamellae [3]. Vascular calcification, including MAC, is a strong predictor of

cardiovascular morbidity and mortality [98]. Almost no research except one recent study

from our laboratory has been directed at reversing of elastin specific MAC [6, 28]. Our

previous studies showed EDTA might be a promising chelating agent to reverse elastin

calcification both in vitro [28]. One recent clinical trial in the US from 2008 to 2013,

named Trial to Assess Chelation Therapy (TACT), aimed to determine the safety and

effectiveness of EDTA chelation therapy by systemic infusion in individuals with

coronary artery disease [240]. Published results of TACT showed that intravenous

chelation regimen with EDTA modestly reduced the risk of adverse cardiovascular

outcomes but did not specifically highlight EDTA chelation therapy whether or not could

reverse vascular calcification [246].

Systemic chelation therapy using chelating agents such as EDTA or sodium

thiosulfate to reverse calcification requires a high dosage and has side effects such as

hypocalcemia, bone loss and renal toxicity [14, 18, 119]. Due to these reasons and short

half-life of EDTA in circulation, no conclusive evidence is available about reversing

arterial calcification by chelation therapy. Our results in previous chapter with systemic

Page 152: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

134

EDTA therapy did not show reversal of MAC when delivered systemically as intravenous

injection. It also caused loss of blood calcium and increase in urine calcium. We

hypothesize that nanoparticle-based targeted chelating agent delivery could lower the

dosage required and have many other advantages such as improved bioavailability and

sustained release of drugs [29]. We have recently shown that the elastin antibody coated

nanoparticles can be targeted to vascular calcification sites [30].

Albumin based nanoparticles (NPs) have been shown as a promising nontoxic,

non-immunogenic drug carrier [183]. Nanoparticle protein engineering therapy using

albumin NPs offers potential solutions to many of the problems like toxicities associated

with the solvent-based formulations [247].

Thus, in this study we tested if albumin NPs may be a good EDTA carrier to

deliver EDTA to the calcification sites. By surface coating with anti-elastin antibody, we

show reversal of elastin specific MAC in vitro or in vivo without side effects associated

with systemic EDTA chelation therapy.

7.2 Materials and Methods

Preparation of EDTA loaded albumin NPs

Albumin NPs were prepared by ethanol desolvation methods [183, 186, 248].

Briefly, 200 mg bovine serum albumin was dissolved in 4 mL of distilled and deionized

water (DD water) and then the pH was adjusted to 8.5 with 6N NaOH. EDTA was added

to this solution at various concentrations (25, 50, 100 or 200 mg) for loading optimization

experiments. The aqueous solution was added drop-wise to 16 mL ethanol under probe

Page 153: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

135

sonication (20 Watts, Omni Ruptor 400 Ultrasonic Homogenizer, Omni International Inc,

Kennesaw, GA) to prepare NPs. The particles were stabilized by fixation with 4%

glutaraldehyde (100 μL) for 1 hour and washed 3 times with DD water. The prepared

NPs were then lyophilized and stored at 4 ℃ for further use. For NP tracking

experiments, NPs were loaded with a fluorescent dye (2.5 mg, DiR, 1, 1-dioctadecyl-3, 3,

3, 3-tetramethylindotricarbocyanine iodide, Biotium, Inc., Hayward, CA) instead of

EDTA.

Characterization of EDTA loaded albumin NPs

Particle size and Zeta potential in suspension was determined using 90Plus

Particle Size Analyzer (Brookhaven Instruments Co, Holtsville, NY). 10 µl of

nanoparticle suspension was diluted in 3 ml of HPLC-grade water in a disposable plastic

cuvette. Particle size was also measured by transmission electron microscope (TEM,

H7600, Japan). Briefly, 20 µl of nanoparticle suspension (0.1 mg/ml) was added on a

formvar-coated copper grid and allowed to be dried overnight under reduced vacuum,

and then copper grid was loaded into the sample chamber of the TEM.

EDTA loading efficiency:

The drug loading efficiency was calculated using the formula:

Loading efficiency (%) = (

) ⑴

The amount of entrapped EDTA was back calculated by subtracting the amount of

non-entrapped EDTA in NPs mixture after separation of synthesized EDTA loaded

Page 154: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

136

albumin NPs. The supernatant wash solution mentioned above during NPs preparation

was collected for calculation of non-entrapped EDTA amount. EDTA was analyzed

based on a standard curve using EDTA-FeCl3 complex by Ultraviolet–visible(UV-Vis)

spectroscopy with peak absorption at 257 nm[249].

In vitro EDTA release

In vitro release studies were performed by incubating 2 mg EDTA loaded albumin

NPs nanoparticles in 1ml of 0.1M Phosphate buffered saline (PBS) at pH 7.4. The NP

suspension was shaken continuously at 37 ℃. At pre-selected times (2, 4, 8, 24, 48, 72,

96 and 120 hours), the supernatant was collected for EDTA quantification.

Determination of cell toxicity and uptake of NPs

For cellular cytotoxicity concentrations of 100, 1000 µg EDTA loaded albumin

NPs /ml cell culture media were used to evaluate possible toxicity to rat aortic smooth

muscle cells (RAMSCs, passage 6). Briefly, RAMSCs were seeded at 10,000 cells/cm2.

When reached at 70% confluency, RAMSCs were incubated with nanoparticles for 4

hours. Then cell viability was determined using a LIVE/DEAD Cell Viability assay

(Molecular Probes, Grand Island, NY). The live controls are cells cultured on normal

media while dead controls are cells treated by 70% ethanol. Cells that have green

fluorescence are considered alive while cells that have red fluorescence are considered

dead in this stain.

Page 155: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

137

Cellular cytotoxicity was also evaluated using MTT (3-(4,5-Dimethyl-2-

thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay. Briefly, 5 mg of MTT (Sigma

Aldrich, St.Louis, MO) was dissolved in 10 ml of PBS and then were added to RAMSCs.

After 4 hours, PBS was carefully aspirated and the insoluble formazan dye was collected

with dimethyl sulfoxide (DMSO) (Sigma Aldrich, St. Louis, MO). Absorbance was read

at 560 nm and normalized to control (normal media) readings.

To visualize cellular uptake of NPs (After antibody coating, described later), NPs

with same concentrations of 100 µg NPs/ml media and 1000 µg NPs /ml media were

tested. DiR dye (Biotium, Inc., Hayward, CA) was used to label the NPs. 24 hours after

DiR-labeled NPs’ incubation, the cells were thoroughly washed with PBS three times to

remove unbound nanoparticles, fixed with 4 % formaldehyde, labeled with the lipophilic

membrane stain DiI (or 1,1'-Dioctadecyl-3,3,3',3'-Tetramethylindocarbocyanine

Perchlorate, Invitrogen, Carlsbad, CA). Imaging of cells was performed using fluorescent

microscopy (EVOS fl. Microscope, Advanced Microscopy Group, Bothell, WA).

Demineralization of calcified tissue in vitro

Calcified porcine aortic elastin (160~180 µg Ca/mg tissue) was prepared by

subdermal implantation of pure porcine aortic elastin in rats as described in earlier

publication [200].

Calcified human aortas (100~300 µg Ca/mg tissue) from cadavers (age 49-75,

both male and female with moderate or severe atherosclerosis) were received from the

Page 156: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

138

Greenville Hospital System (Greenville, SC) following standard dissection technique. All

samples were from the abdominal aorta, just below the renal arteries.

To determine EDTA loaded albumin NPs’ efficacy to remove calcium from

calcified tissue, calcified porcine aortic elastin and separately calcified human aorta (10

mg each) were soaked in 10 ml EDTA loaded albumin NPs dispersion (10 mg/ml in PBS,

n=3) with constant shaking at 37 ℃. The control groups were soaked in 10 ml blank

albumin NPs without EDTA loading (10mg/ml in PBS, n=3). 3 days after treatments all

tissue samples were embedded in paraffin blocks, sectioned, and stained for qualitative

analysis of calcium with Dahl’s alizarin red stain.

Antibody coated EDTA loaded albumin NPs – preparation and optimization for antibody

binding

The antibody surface coating strategy for albumin NPs is illustrated in Figure

7.1A. Antibody thiolation: 10 µg of antibodies were thiolated using 34 µg of freshly

prepared Traut’s reagent (G-Biosciences, Saint Louis, MO) in 0.1 ml of PBS for 1 hour.

Antibodies used include Alexa-Fluor 555 goat anti-mouse IgG (Molecular Probes,

Carlsbad, CA), rabbit anti-rat IgG antibody (Thermo Scientific, Rockford, IL) and rabbit

anti-rat elastin antibody (United States Biological, Swampscott, MA). Thiolated

antibodies were washed three times with PBS and dialyzed through 10 kDa MWCO

filters to remove Traut’s reagent.

Albumin NPs conjugate to Maleimide-PEG- NHS ester: EDTA loaded albumin

NPs were prepared as described earlier. Albumin NPs were activated with

Page 157: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

139

heterobifunctional crosslinker α-maleimide-ω-N-hydroxysuccinimide ester poly (ethylene

glycol) (Maleimide-PEG- NHS ester, MW 2000 Da, Nanocs Inc.,NY, USA) in order to

achieve a sulfhydryl-reactive particle system. 2.5 mg of Maleimide-PEG- NHS ester

solution was added into 10 mg albumin NPs dispersion. Then the mixture was left under

shaking for 1 hour at room temperature.

Albumin NPs conjugate to antibodies: purified and thiolated antibodies were then

added to the activated albumin NPs and incubated overnight at room temperature for

conjugation. Samples were centrifuged and the supernatant was analyzed using

fluorescent spectrometry (n=3). A calibration curve was plotted from standard solutions

(0.1-25µg of Alexa-Fluor 555 goat anti-mouse IgG in 1 ml of PBS). The bound dye was

back calculated by subtracting the amount of unbound dye in the supernatant.

Page 158: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

140

Page 159: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

141

Figure 7.1. A: Schematic representation of antibody surface coating strategy for albumin

NPs. 1) Antibody thiolation using traut’s reagent to produce a terminal sulfhydryl group

on antibody. 2) Albumin NPs conjugated to NHS ester-PEG-Maleimide with amide bond.

3) Antibody conjugated to albumin NPs with maleimide – sulfhydryl reaction. B:

Experimental protocol for CaCl2 injury model and treatments of rats. CaCl2 injury model

(represented as a symbol of arrows) was created at starting time point of experiment (Day

0, week 0). There were total five treatment groups with following treatments by tail vein

injection (two times a week, two weeks): 1) saline as control group (n=5, 5 rats); 2) High

dose (HD) EDTA group (n=5, 5 rats, 50 mg/kg EDTA); 3) Low dose(LD) EDTA group

(n=3, 3 rats, 2.5 mg/kg EDTA); 4) IgG-NPs/EDTA group (n=5, 5 rats, 5 mg of NPs for

each rat at each injection); 5) EL-NPS/EDTA group (n=5, 5 rats, 5 mg of NPs for each rat

at each injection). Rats were placed in metabolic cages for urine collection for 24 hours

(Day 21); then all rats were given another injection on Day 22 (depending on individual

treatment groups) and then urine samples were taken at time points of 1, 2, 3 and 4 hours

post injection; right after that rats were euthanized and organs were harvested

(represented as a symbol of six-point stars).

Immunostaining to visualize antibody coating on NPs

Rabbit anti-rat IgG antibody coated albumin NPs (IgG-NPs) were prepared as

described earlier. 0.1 ml of NPs (1 mg/ml) without antibody coating and IgG-NPs (2 µg

IgG/mg NPs, 1 mg/ml) were incubated with 0.1 ml of 10 nm gold stained goat-anti-

Page 160: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

142

rabbit IgG (Sigma Aldrich, St.Louis, MO) overnight in 0.01M tris buffered saline (TBS),

with 0.05% bovine serum albumin and 15% glycerol and 7.5 mM sodium azide. NPs

were washed twice with TBS to remove unbound antibodies at 14,000 × g for 15 minutes

after incubation. Then 20 µl of NPs’ suspension (1 mg/ml) was added on a formvar-

coated copper grid and allowed to be dried overnight under reduced vacuum. Antibody

surface conjugation was then analyzed by TEM.

Ex-vivo nanoparticle targeting studies using elastase damaged aorta

Aortas from Sprague-Dawley rats (Pel-Freez Biologicals, Rogers, AR) were used

in these studies (n=2, 2 samples in each group, total 8 groups). High purity porcine

pancreatic elastase (Elastin products company, Owensville, Missouri) was prepared

(5U/ml) in 100mM Tris Buffer, 1mM calcium chloride, 0.02% sodium azide, pH 7.8.

Elastase damaged aortas were prepared by injecting elastase intra-luminally for 1 hour to

mimic a damaged aorta in medical calcification [30]. Intra-luminal injection was done by

clamping both ends of the aorta during injection.

Rabbit anti-rat elastin antibody coated albumin NPs (EL-NPs) or rabbit anti-rat

IgG antibody coated albumin NPs (IgG-NPs) loaded with DiR dye were prepared by

following the same method as described earlier, Antibody coated NPs were injected

either in control fresh healthy aortas (without elastase treatment) or with elastase

damaged aortas. Following injection, the aortas were thoroughly washed 3 times with

PBS to minimize non-specific adherence of nanoparticles. The bound NPs (DiR dye

fluorescence) in aortas were quantified by Caliper IVIS Lumina XR image system

Page 161: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

143

(Hopkinton, MA) with Ex/Em of 745/810 nm. The dry weights of the aortas were

recorded for fluorescent density normalization.

In vivo nanoparticle targeting studies based on local periadventitial CaCl2 injury model

CaCl2 injury model was used to mimic local aortic calcification at the abdominal

aortic region following previous protocol [27, 28]. Briefly, 6 male Sprague-Dawley (SD)

rats (6 to 8 weeks old) were anesthetized by 2-3% isoflurane. A ventral laparotomy

exposed the peritoneal cavity. A 15 mm section of the infrarenal abdominal aorta was

exposed and injury was created by placing 0.5 M CaCl2 soaked sterile cotton gauze on

top of the aorta for 15 minutes. After CaCl2 application, the gauze was removed and the

area was briefly flushed with warm saline. Incisions were then closed with continuous

sutures (Abdominal and subcutaneous) and surgical staples. Then rats were allowed to

recover and maintained in standard chow conditions for one week.

After one week (when calcification in injured site is seen), rats were anesthetized

and EL-NPs/IgG-NPs were injected through the tail vein (10 mg of NPs/kg body weight

in 0.1 ml saline). Rats were euthanized 24 hours after the injection.

Only organs that showed fluorescent signal were lyophilized and used to calculate

bio-distribution, which include aorta, heart, lung, liver, kidney, spleen. All other organs

and tissue such as brain, muscle, skin, and blood did not have detectable fluorescent

signal.

Percentage of targeting for each organ was calculated as follows:

Page 162: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

144

%targeting

) / (dry weight of organ))*100

In vivo nanoparticle therapeutic efficacy studies based on local periadventitial CaCl2

injury model

An illustration of experimental design for this animal study was shown in Figure

7.1B. Firstly at starting time point (Day 0) CaCl2 injury was created same as mentioned

above. CaCl2 injury model mimics local elastin specific MAC. One week later rats were

divided into five groups for the following treatments by tail vein injection (two times a

week, two weeks): 1) saline as control group (n=5); 2) High dose (HD) EDTA group

(n=5, 50 mg/kg EDTA); 3) Low dose (LD) EDTA group (n=3, 2.5 mg/kg EDTA); 4)

IgG-NPs/EDTA group (n=5, 5 mg of NPs for each rat at each injection); 5) EL-

NPS/EDTA group (n=5, 5 mg of NPs for each rat at each injection). Rats were placed in

metabolic cages for urine collection for 24 hours at the end of the treatment day (Day 21).

Then all rats were given another injection on Day 22 (depending on individual treatment

groups) and then urine samples were taken at time points of 1, 2, 3 and 4 hours post

injection and were kept in – 80 ℃ for further analysis. Right after that rats were

euthanized by CO2 asphyxiation and organs were harvested including the aorta (from the

heart to the iliac bifurcation), blood, femoral bone, kidney, heart, liver and spleen.

Heparin (10 units/ml blood) was used as anti-coagulant for blood samples and serum was

removed and stored at – 80 ℃ for further assay. Aorta, kidney, heart, liver and spleen

were fixed in 10% buffered formalin for further analysis.

Page 163: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

145

Aortic calcifications were evaluated by stereomicroscope (Leica M80 stereo

microscope, Leica Microsystems Inc. Buffalo Grove, IL), micro-computed tomography

(micro-CT) scanner (Siemens Inveon microPET/CT preclinical scanner, Siemens

Healthcare Molecular Imaging, USA), histology and atomic absorption spectrometry

(AAS). Aortas from all rats were cleaned of all adherent tissues by blunt forceps after

formalin fixation. Plastic tube (PE, 1 mm outside diameter) was inserted into the cleaned

aortas for easier handling during imaging. Whole aortas were imaged by

stereomicroscope (Leica M80 stereo microscope, Leica Microsystems Inc. Buffalo Grove,

IL) both before and after alizarin red staining [96, 243, 244]. Briefly, whole aortas were

soaked in freshly made 2% alizarin red solution (pH 4.1-4.3, pH) for 10 minutes and

washed by distilled and deionized water for another 5 minutes. Then aortas were imaged

by stereomicroscope. Micro-CT for harvested aortas in vitro was based on modified

protocol [237, 239]. Briefly, a 360-degree axial scan was collected at a peak tube

potential of 80kVp and tube current of 500uA. Data were reconstructed to 100 μm pixel

size using a Feldkamp cone beam algorithm. Virtual 2D cross-sections and 3D maximum

intensity projection views were created using MeVisLab software (MeVisLab, Mevis,

Bremen, Germany, http://www.mevislab.de). Exported movies from MeVisLab were

further edited by video-editing software- Corel VideoStudio Pro X7(Corel Inc, Mountain

View, CA) (Available as online video supplement 4).

Following the non-invasive imaging by stereomicroscope and micro-CT scanner

mentioned above, aortas were processed for histology with alizarin red staining for

visualization of calcium. Remaining parts of abdominal aortas were then used for Ca

Page 164: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

146

quantification by atomic absorption spectroscopy, AAS [28, 232]. Briefly, 2 ~ 10 mg of

abdominal aortas ware hydrolyzed in 1ml of 6N Ultrex HCl (Baker Company,

Phillipsburg, NJ) and then re-suspended in 0.01N Ultrex HCl. Hydrolyzed samples were

diluted and analyzed with AAS. Serum and urine samples were thawed and diluted

before Ca quantification by AAS.

Rats’ femoral bones were dissected and fixed in 10% buffered formalin. The

integrity of femoral bone was evaluated by Micro-CT and by functional mechanical test.

Stiffness and maximum load were tested based on three points bending test. Micro-CT

for rats’ femoral bones was done by same protocol as aortas mentioned earlier (One

representative scanning data for the whole 2D view slices from each group is available as

online supplement video 5). Three points bending test was done using Bose test machine

(Electroforce 3200, Bose, MN, USA) equipped with a 450N load cell. First, stiffness was

tested by measuring bone displacement (in Millimeter, mm) at a given force (in Newton,

N). Second, bones were exposed to higher force until fracture occurred and the maximum

enduring load (N) was determined.

Statistical analysis

Results are expressed as means ± standard deviation (SD). Statistical analyses of

the data were performed using single-factor analysis of variance. Differences between

means were determined using the least significant difference with an α value of 0.05.

Asterisks in Figures indicate statistical significance (P < 0.05).

Page 165: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

147

7.3 Results

Characterization for EDTA loaded albumin NPs – optimization and function

The size of NPs was dependent on sonication time; 10, 45 and 60 minutes of

sonication resulted in 1207.9 ± 177.3, 144.1±10.6 and 121.9 ± 13.5 nm size NPs

respectively (Figure 7.2A, 7.2B). Studies have shown that particle size with ~200 nm

could get sufficient retention of nanoparticles in the extracellular matrix and also

minimum cellular uptake by VSMCs [182, 250, 251]. So all NPs used later were prepared

by 45 minutes of probe sonication to get particles with ~200 nm size. Zeta potential of

NPs after antibody modification is shown in Table 7.1. As we wanted to target

extracellular matrix elastin and reduce cellular uptake, all NPs used in the animal study

were negatively charged (due to the inherent negative charge on the mammalian cell

membrane, negatively charged particles are repelled) (Table 7.1). EDTA loading

increased with initial EDTA concentration used but plateaued to ~ 22 % with no further

increase in loading with additional starting EDTA concentration (Figure 7.2 C). Release

profile for EDTA loaded albumin NPs showed initial burst and then continuous release

for up to 5 days (Figure 7.2 D) both before and after antibody modification. The amount

of total EDTA was slightly lower after antibody modification because of loss during

washing steps in antibody coating.

EDTA loaded albumin NPs did not cause cytotoxicity to RASCMs based on

Live/Dead assay and MTT assay in the range of 100 µg NPs/media to 1000 µg

NPs/media (Figure 7.2 E and 7.2 F). EDTA loaded albumin NPs did not show cellular

Page 166: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

148

uptake in the concentration range between 100 µg NPs/media to 1000 µg NPs/media

(Figure 7.2 G). EDTA loaded albumin NPs could remove calcified mineral efficiently

both from calcified elastin and calcified human aorta samples as shown by Alizarin red

stain (Figure 7.2 H). The blank albumin NPs without EDTA loading were unable to

regress calcification (Figure 7.2 H).

Table 7.1. Characterization of nanoparticles.

Type of

nanoparticle

Size ±

SEM(nm)

Poly dispersity

index

ζ- potential ± SD

(mV)

IgG-NPs/DiR 177.9 ± 21.6 0.795 - 22.89 ± 5.28

EL-NPs/DiR 144.1± 10.6 0.479 - 31.72 ± 3.44

Page 167: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

149

Page 168: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

150

Figure 7.2. Characterization for albumin NPs – optimization and function. A: Particle

size measured by 90Plus Particle Size Analyzer with sonication times of 10, 45, and 60

minutes. B: Images showed particle size measured by TEM with corresponding

sonication times. C: Loading efficiency with serials of EDTA and albumin ratios. D:

Release profile for EDTA loaded albumin NPs before and after antibody modification.

Ab: antibody. E: LIVE/DEAD cell viability assay. Live control - normal media; Dead

control - treated by 70% ethanol; 100 µg NPs/ml media and 1000 µg NPs /ml media are

two studies groups. Magnification: 100X. Scale bar: 400µm. F: Cell viability determined

by MTT assay. G: Cellular uptake study with two NPs’ concentrations of 100 µg NPs/ml

media and 1000 µg NPs /ml media. Red fluorescence: cell membrane stained by

lipophilic membrane stain DiI (Excitation/Emission: 549/565nm). Purple fluorescence:

albumin NPs stained by DiR near-infrared dye (Excitation/Emission: 745/810 nm).

Magnification: 100X. Scale bar: 400µm. H: Demineralization of calcified elastin and

calcified human aorta. Blank albumins NPs without EDTA were control groups. EDTA

loaded albumin NPs were study groups. Alizarin red stains calcium as red and

counterstained by light green. Magnification: 100X. Scale bar: 100 µm.

Antibody coated EDTA loaded Albumin NPs – antibody binding and targeting efficacy

in vitro

Antibody binding yield was assessed by Alexa-flour 555 labeled IgG based on

standard curve (data not shown) of Alexa-fluor 555 conjugated IgG with

excitation/emission: 555/595 nm. The amount of IgG bound to albumin NPs increased

with more starting amount of antibody added but the bound percentage peaked around 5

µg IgG / mg albumin NPs (Figure 7.3 A and 7.3 B). Successful antibody coating was

also verified by gold immune-staining under TEM showing 10 nm gold particles on the

surface in the IgG-NPs (2 µg/mg IgG-NPs) while no gold particles presented in the

control NPs without antibody coating (Figure 7.3 C).

Control healthy aortas without NPs treatment showed no auto-fluorescence

(Figure 7.3 D); Control healthy aortas without elastase damage showed little

Page 169: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

151

fluorescence. This indicated that either IgG-NPs or EL-NPs would not bind to healthy

aortas. Only damaged aortas (with antibody concentration of 1, 2.5, 5 and 10 µg/mg EL-

NPs) had strong fluorescence. The relative percentages of EL-NPs bound to damaged

aortas were normalized to control samples (1 µg/mg IgG-NPs). Fluorescent density was

quantified based on IVIS fluorescent counts (Figure 7.3E) and showed that antibody

concentration in range of 1 to 5 µg/mg EL-NPs could increase NPs binding to 159.8 % (1

µg/mg EL-NPs) or 176.1% (5 µg/mg EL-NPs). 2.5 µg/mg EL-NPs had good targeting

efficacy (Figure 7.3E) and higher ratio of antibody to NPs did not significantly increase

targeting efficacy so this ratio was chosen for later animal studies.

Figure 7.3. Antibody binding and targeting efficacy in vitro. A: Amount of IgG antibody

bound in 1 mg of albumin NPs with serials of starting ratio of antibody and albumin NPs

(1, 5, 10, 25 µg IgG/mg NPs). B: Percentage of IgG antibody bound with albumin NPs

Page 170: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

152

with serials of starting ratio of antibody and albumin NPs (1, 5, 10, 25 µg IgG/mg NPs).

C: Gold immune-stained albumin NPs by TEM. Left: NPs without antibody coating

serviced as negative control. Right: NPs with IgG antibody coating, antibody

concentration used: 2 µg/mg IgG-NPs. Small black dots on NPs’ surface (right) represent

10 nm gold stained IgG. D: Fluorescent image for aortas by IVIS image system. Aortas

of numbers 1 to 8 are controls. Aortas of numbers 9 to 16 are study groups. Samples 7 to

16 in red rectangle area were used for DiR quantification. E: Quantification of EL-NPs

bound percentage by fluorescent density from IVIS images. C in horizontal axle: control

groups of sample 7 and 8(Damaged aortas and treated with 1 µg/mg IgG-NPs).

Fluorescent density of study groups (aortas numbers 9 to 16) were normalized to control

groups of sample 7 and 8.

Efficacy of EL-NPs targeting to damaged aorta in vivo

Most of the DiR dye was entrapped permanently in albumin NPs (data not shown),

thus NP tracking with fluorescence was possible for bio-distribution in animal studies.

Photography, fluorescence and X-ray image for all six aortas has been shown in Figure

7.4A. Red rectangle areas show where injury caused elastin damage and calcification in

the abdominal aortas. Those areas showed considerably more fluorescent signal in the

study groups (treated with EL-NPs/DiR) and no fluorescence for IgG-NPs/DiR.

Representative histological sections from damaged aortas both from EL-NPs/DiR and

EL-NPs/DiR treated groups showed that EL-NPs had better targeting efficacy and could

infiltrate from lumen to adventitia in damaged blood vessel (More purple fluorescence

from DiR dye labeled as red arrows in Figure 7.4B). Green fluorescence was auto-

fluorescence from aortic tissue section. EL-NPs treated groups had more NPs distribution

in aortas (15.98% in EL-NPs versus 5.75% in IgG-NPs) while less in spleen, kidney, lung

and heart comparing to control IgG-NPs treated groups (Table 7.2).

Page 171: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

153

Figure 7.4. Efficacy of EL-NPs targeting to damaged aorta in vivo. A: Photography,

fluorescent image and X-ray image taken by IVIS image system. IgG-NPs/DiR: rabbit

anti-rat IgG antibody modified albumin NPs entrapped with DiR dye. EL-NPs/DiR:

rabbit anti-rat elastin antibody modified albumin NPs entrapped with DiR dye. The red

rectangle areas were calcified and damaged region detected by X-ray. B: Fluorescent

microscopy of abdominal aorta from representative histological section. Purple

fluorescence: albumin NPs stained by DiR near-infrared dye (Excitation/Emission:

745/810 nm). Green fluorescence: auto-fluorescence from aortic tissue section. Red

arrows indicated area with more purple fluorescence. Left: aortic section from IgG-

NPs/DiR groups as control. Right: aortic section from EL-NPs/DiR groups which

indicated that EL-NPs had better targeting efficacy and could infiltrate from lumen to

adventitia in damaged blood vessel.

Page 172: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

154

Table 7.2. Organ distribution of fluorescent nanoparticles.

Organ % total fluorescence/g dry weight

(IgG-NP/DiR)

% total fluorescence/g dry weight

(EL-NP/DiR)

Aorta 5.75 ± 0.54 15.9 8 ± 0.87

Liver 32. 47 ± 0.83 36.60 ± 8.44

Spleen 35.55 ± 3.55 30.84 ± 5.48

Kidne

y 14.38 ± 1.81 11.59 ± 2.77

Lung 6.76 ± 0.23 3.37 ± 0.49

Heart 5.10 ± 3.18 1.61 ± 0.30

Therapeutic effects of targeted EDTA chelation therapy to reverse calcification

Aortic calcification for both systemic EDTA chelation therapy and targeted

EDTA chelation therapy is shown in Figure 7.5. Aortic calcification evaluated by

stereomicroscope before and after whole aorta’s alizarin red stain, Micro CT, and

histological stain by alizarin red showed that EL-NPs/EDTA groups showed best

therapeutic effects to reverse aortic calcification (Figure 7.5A). Four out of five aortas

showed complete reversal of calcification. No other groups including IgG NPs, low and

high EDTA systemic injections showed any significant reduction in calcification of the

aorta (Figure 7.5 A). These staining results corroborated calcium quantification in aorta

by AAS. Quantitative calcium results also showed that EL-NPs/EDTA groups had best

Page 173: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

155

therapeutic effect to reverse aortic calcification and had lowest Ca amount in the aorta

(Figure 7.5B, P = 2.63E-06, n =5).

Page 174: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

156

Page 175: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

157

Figure 7.5. Evaluation of aortic calcification for both systemic EDTA chelation therapy

and targeted EDTA chelation therapy. A: Aortic calcification evaluated by

stereomicroscope before and after whole aorta’s alizarin red stain, Micro CT and

histological stain by alizarin red. Here showed results of IgG-NPs/EDTA and EL-

NPs/EDTA groups. B: Ca quantification in aorta by AAS for all five treatment groups

(For IgG-NPS/EDTA and EL-NPs/EDTA groups, P = 2.63E-06, n =5).

Targeted EDTA chelation therapy did not have side effects associated with systemic

EDTA chelation therapy

Possible side effects with both systemic EDTA chelation therapy and targeted

EDTA chelation therapy are shown in Figure 7.6. Results showed no side effects to

serum and urine Ca as seen in high dose EDTA treated groups in EL-NPs/EDTA groups,

or targeted EDTA chelation therapy groups, (Figure 7.6A, P = 0.0045, n = 5; Figure

7.6B, P = 0.0001, n = 5; Figure 7.6C). Similarly, there were no bone losses seen in all

treated groups both from morphology and from functional test of stiffness and fracture

stability (Figure 7.6D; 7.6E; 7.6F, P = 0.027, n =3).

Page 176: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

158

Page 177: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

159

Figure 7.6. Possible side effects with systemic EDTA chelation therapy and targeted

EDTA chelation therapy. A: Serum Ca quantification for all five treatment groups (For

Saline and HD EDTA groups, P = 0.0045, n = 5). B: Urine Ca quantification for all five

treatment groups (For Saline and HD EDTA groups, P = 0.0001, n = 5). C: Urine kinetic

Ca quantification after tail vein injection of all five different reagents up to four hours. D:

Bone morphology analyzed by Micro CT showing 15 mm below the trochanter major of

femoral bone for all five treatment groups. E: Stiffness of femoral bone for all five

treatment groups. F: Maximum load of femoral bone for all five treatment groups (For

Saline and HD EDTA groups, P = 0.027, n =3).

7.4 Discussion

To our knowledge this is the first time anyone has shown that targeted

nanoparticles when injected systemically can bind to calcified artery and regress

calcification in vivo. Vascular calcification occurs at two different sites- intima and media.

Page 178: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

160

Intimal calcification is generally associated with atherosclerotic plaques and

inflammation and can occlude the lumen while medial calcification occurs in the elastin

lamina of medium and large arteries in patients with chronic kidney disease (CKD) or

diabetes causing increase in arterial stiffness. Surgical methods like directional

atherectomy and stent grafts are used to treat atherosclerosis and intimal calcification and

to open occluded arteries [4, 5]. However, there are no treatments available for medial

elastin-specific calcification.

Chelation therapy has been touted to remove calcification of arteries, mostly for

intimal atherosclerotic calcification; however, very few animal studies have been

performed and none of the clinical studies have proven its effectiveness in improving

cardiovascular function [174, 176]. We wanted to test if elastin-specific medial

calcification can be removed by targeted chelation therapy. We chose EDTA as our

previous comparative study of three chelating agents: EDTA, sodium thiosulfate and

DTPA (diethylene triamine pentaacetic acid) showed that EDTA worked best to remove

calcium from hydroxyapatite and calcified tissue [28]. In the same study, we have also

shown that local placement of EDTA loaded poly(lactic-co-glycolic) acid (PLGA),

nanoparticles on top of calcified aorta after exposing calcified abdominal aorta by ventral

laparotomy, could effectively reverse elastin-specifically medical calcification in a rat

calcification model [28]. However, such invasive procedure cannot be translated for

human use. In an attempt to target drugs to diseased arteries, we recently discovered that

specifically designed NPs with elastin-targeting antibodies on the surface were able to

deliver agents to the site of elastic-lamina damage [30]. Elastin fibers in elastic lamina in

Page 179: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

161

healthy artery are covered with microfibrillar glycoproteins and thus not accessible to

elastin antibody targeted to elastin. In most vascular diseases, elastic lamina gets

damaged and elastic fibers get exposed. We used this knowledge to create NPs that

specifically targeted to diseased site in artery [30]. These previous studies paved the way

to design an elastin antibody coated and EDTA loaded albumin NPs to reverse elastin-

specific MAC and to avoid side effects associated with systemic EDTA chelation therapy.

We choose albumin nanoparticles instead of polymeric nanoparticles like PLGA or PLA

(Polylactic acid) because albumin NPs are less toxic as no organic solvents are used in

preparation and used frequently in clinical applications [183, 247]. We could also load

significant amount of EDTA in albumin NPs rather than PLGA NPs.

We optimized surface charge and size of albumin NPs so that they are not taken

up by cells and remain in the extracellular matrix. Others have shown that small particle

size less than 100 nm would have more uptake in vascular smooth muscle cells [250]

while big particles larger than 500 nm could not effectively penetrate endothelium and

basement membrane in blood vessels. Previous results in our lab indicated that

nanoparticles of ~200 nm size were able to penetrate both the endothelium and the

basement membrane and also limited cellular uptake [30]. The size of EDTA loaded

albumin NPs was quite dependent on the duration of sonication time and we found that

45 minutes of probe sonication resulted in 150~200 nm size. Since mammalian cell

membrane has negative charge, negatively charged particles show less cellular uptake

[252]. Our result showed that our negatively charged albumin NPs with size of 150~200

nm have limited cellular uptake. We could load up to 20 % EDTA (wt/wt) and NPs

Page 180: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

162

sustained release of EDTA for up to 5 days. We found EDTA loaded albumin NPs did

not cause significant cell death or uptake. Next we checked if such NPs can remove

calcium from calcified elastin and human aorta in vitro. Indeed, EDTA releasing NPs

when incubated with calcified tissues removed calcium both from calcified elastin and

calcified human aorta in vitro. Once the charge, size, and targeting and therapeutic

efficiency was tested in vitro , we next designed in vivo experiments to test if such NPs

can deliver EDTA to the calcified aorta under high shear and blood flow present in the

aorta. We injected NPs through rat tail vein and allowed them to circulate and target to

the site. We clearly found that NPs (with DiR dye) with elastin antibody on the surface

(EL-NPs/DiR) did accumulate at the inured aortic site while IgG-NPs with DiR did not.

We then injected NPs containing chelating agent EDTA with either elastin antibody on

the surface (EL-NPs/EDTA) or irrelevant IgG antibody (IgG-NPs/EDTA). We decided to

inject NPs 2 times a week based on in vitro release profile for EDTA. Within 2 weeks of

injections (total 4 injections) we could observe complete removal of calcium from 4 out

of 5 arteries in EL-NPs EDTA group as confirmed by alizarin red staining, microCT, and

quantitative calcium scores. Not only calcium was removed but the elastic lamina seemed

to be less damaged after removal of calcium. On the other hand EDTA loaded IgG NPs

did not target to the site and did not remove any calcium. We also injected EDTA

systemically in the same manner NPs were delivered (2 times a week), either in high dose

(similar to what is used clinical studies) or low dose (based on how much total EDTA

was delivered through NPs). In both of these direct injection cases, EDTA failed to

remove calcium from the arteries as there was no change in calcification seen by alizarin

Page 181: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

163

red staining, quantitative calcium results, and microCT experiments. Moreover, we found

that EDTA dose similar to what used clinically (50 mg/kg) caused significant adverse

events for serum and urine calcium, specifically hypocalcemia and hypercalciuria. Our

NPs contained 20 times lower EDTA (which was protected) and it was released slowly

over a period of five days. Thus, we did not find any systemic toxicity for nanoparticle

groups. Thus, elastin antibody coated and EDTA loaded albumin NPs might be a

promising nanoparticle protein engineering therapy to reverse elastin-specific medial

calcification. This targeted nanoparticle delivery method will lower the dosage required

for chelating agent - EDTA so as to avoid side effects associated with systemic

intravenous infusion such as hypocalcemia and bone loss [14, 18, 119]. It is a safe and

non-invasive method to reverse calcification compared to surgical methods. Antibody-

mediated tissue targeting of albumin NPs for clinical practice has been approved by the

FDA for other drugs [253, 254] so our antibody coated albumin NPs have great potential

clinical relevance.

Limitations of Our Study: There are some limitations to our studies. First, our

EDTA chelation therapy was performed for a period of only two weeks. We do not know

if calcification will return after halting the therapy and also we do not know long the NPs

will stay at the of calcification after intravenous injection. Secondly, we did not

investigate possible side effects besides serum calcium, urine calcium, and bone loss.

More kidney functional evaluation might be necessary such as serum creatinine level.

Finally, we only evaluated EDTA chelation therapy in rat CaCl2 injury model, which

induces local medial calcification at the site of injury. Patients have calcification

Page 182: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

164

dispersed throughout arterial tree thus calcification animal models such as warfarin

[96]or vitamin D [99] application where calcification is seen at various places such as

aortic arch and renal and femoral bifurcation might be necessary in order to investigate

EDTA chelation therapy’s therapeutic effects. Careful risk-benefit analysis needs to done

before use in individual human patients.

7.5 Conclusions

We prepared EDTA loaded albumin NPs with optimized size of 150~200 nm,

zeta potential of – 22.89 ~ - 31.72 mV, loading efficiency of 15~ 20 %, and a sustained

release of EDTA for up to 5 days. Both ex-vivo study and in vivo study using aortic

calcification model showed that elastin antibody coated and EDTA loaded albumin NPs

could have good targeting efficacy and also therapeutic effects to reverse elastin-specific

medial calcification. EDTA loaded albumin NPs did not cause side effects of serum and

urine Ca, as seen in high dose EDTA treated groups. There was no bone loss in all treated

groups. Elastin antibody coated and EDTA loaded albumin NPs might be a promising

nanoparticle protein engineering therapy to reverse calcification in elastin-specific medial

calcification and to avoid side effects associated with traditional EDTA chelation therapy

by systemic intravenous infusion.

Page 183: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

165

Supplemental figures

Page 184: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

166

Figure 7.7. H&E staining for rats’ kidneys (All five groups). No treatment induced

histological change was found.

Page 185: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

167

Figure 7.8. Flow chart for preparation of EDTA loaded albumin NPs.

Page 186: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

168

Figure 7.9. Graphical abstract to show nanoparticle structure and targeting and chelation.

A: nanoparticle schematic. EDTA (yellow) was encapsulated into albumin nanoparticles

(NPs, green). EDTA was chelating agent to chelate calcium complex deposition along

elastic lamina fibers. NHS ester-PEG-Maleimide was bifunctional spacer. NHS ester

formed a stable amide bond through amine group on albumin. Reaction of a sulfhydryl on

antibody (blue) to the maleimide group results in formation of a stable thioether linkage.

B: NPs targeting and chelation schematic. Elastic fibers are formed by 90% mature cross-

linked elastin core and 10% microfibrils (arrows) components. In disease state

mircofibrils degrade and calcium complex depositions occur along elastic lamina fibers.

Exposed elastin allows elastin antibody coated albumin NPs (EL-NPs) targeting and

released EDTA chelates calcium complex depositions (purple) eventually reverse elastin-

specific medial calcification.

Page 187: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

169

Figure 7.10. DiR standard curve and release profile in vitro.

Methods for figure 7.9: Characterization of antibody coated EDTA loaded albumin

NPs - DiR release profile in vitro

Page 188: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

170

A DiR standard curve was plotted using a serial of standard solution (0, 0.05, 0.10, 0.15,

0.25 mg DiR/ml in DD water) by fluorescent spectrometry (Excitation/Emission of

745/810 nm). 5 mg of NPs (n =3, prepared as described earlier, after antibody coating)

were soaked in 1ml of PBS at pH 7.4. The NPs suspension was continuously shaken at

37 ℃. At pre-selected times (2, 4, 6, 8, and 24 hour), The supernatant was collected for

DiR quantification. After 24 hours the NPs were homogenized by Powered 125

homogenizer (Fisher Scientific, MA) and Omni Ruptor 400 Ultrasonic Homogenizer

(Omni International Inc, Kennesaw, GA) to extract non-released DiR in PBS. The

supernatant was collected to detect entrapped DiR after 24 hours. Our data showed that

DiR release was less than 30% (data not shown) within in 24 hours. Most of the DiR dye

would still be entrapped in albumin NPs after 24 hours. So the near-infrared fluorescent

signal from DiR dye could be used to track the albumin NPs bio-distribution in animal

studies.

Page 189: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

171

Figure 7.11. NPs bio-distribution present by organs. A summary of all 6 rats’ NPs

distribution based on near-infrared fluorescent signal based on DiR dye (Ex: 745 nm, Em:

810 nm/ICG).

Page 190: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

172

Figure 7.12. NPs bio-distribution present by individual rats. A summary of all 6 rats’

NPs distribution based on near-infrared fluorescent signal based on DiR dye (Ex: 745 nm,

Em: 810 nm/ICG).

Page 191: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

173

Figure 7.13. One representative aorta’s Micro CT images from all five groups.

Page 192: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

174

CHAPTER 8: CONCLUSIONS AND RECOMMENDATIONS

8.1 Conclusions

The broad goal of this project was to understand mechanisms of elastin specific

MAC, to evaluate efficacy of chelating agents to reverse elastin specific MAC, to

evaluate therapeutic effects of systemic disodium ethylene diaminetetraacetic acid

(EDTA) chelation therapy in the aspect of reversal of calcification and possible side

effects, and to develop a targeted delivery system to reverse elastin specific MAC and to

avoid side effects.

We show the following:

1) In aim 1, we demonstrate that RASMCs lose their smooth muscle lineage markers

like alpha smooth muscle actin (SMA) and myosin heavy chain (MHC) and undergo

chondrogenic/osteogenic transformation in presence of mineral deposits. This is

indicated by an increase in the expression of typical chondrogenic proteins such as

aggrecan, collagen type II alpha-1 (Col2a1) and bone proteins such as runt-related

transcription factor 2 (RUNX2), alkaline phosphatase (ALP) and osteocalcin (OCN).

Furthermore, when calcified conditions are removed, cells return to their original

phenotype. Our data supports the hypothesis that elastin degradation and calcification

precedes VSMCs’ osteoblast-like differentiation.

2) In aim 2, we show that both EDTA and diethylene triamine pentaacetic acid (DTPA)

could effectively remove calcium from HA and calcified tissues, while sodium

thiosulfate (STS) was not effective. The tissue architecture was not altered during

Page 193: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

175

chelation. In the animal model of aortic elastin-specific calcification, we further

show that local periadventitial delivery of EDTA loaded in to poly (lactic-co-glycolic

acid) (PLGA) nanoparticles regressed elastin specific calcification in the aorta.

Collectively, the data indicate that elastin-specific medial vascular calcification could

be reversed by chelating agents.

3) In aim 3, we demonstrated that systemic EDTA chelation therapy did not reverse

local aortic calcification. Systemic EDTA chelation therapy caused adverse events

for serum and urine calcium, specifically hypocalcemia and hypercalciuria, but did

not cause bone loss or affect kidney function may be due to short treatment times of

two weeks. These results indicated that systemic EDTA chelation therapy might not

have therapeutic effects in the aspect of reversal of calcification and a strict protocol

is required for safety dosage.

4) In aim 4, we developed EDTA loaded albumin Nanoparticles (NPs) that could

release EDTA slowly. We prepared EDTA loaded albumin NPs with optimized size

of 150~200 nm, zeta potential of – 22.89 ~ - 31.72 mV, loading efficiency of 15~ 20

%, and a sustained release up to 5 days. We further show that when such NPs were

decorated on the surface with elastin antibody, they could be targeted to degraded

elastic lamina efficiently when administrated by intravenous injection. Both ex-vivo

study and in-vivo study using local aortic calcification model showed that elastin

antibody coated and EDTA loaded albumin NPs have good targeting efficacy and

also therapeutic effects to reverse elastin-specific medial calcification. Such NPs

delivery did not cause systemic side effects that were seen for direct injection of

Page 194: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

176

EDTA. Elastin antibody coated and EDTA loaded albumin NPs might be a

promising nanoparticle protein engineering therapy to reverse calcification in elastin-

specific medial calcification and avoid side effects associated with traditional EDTA

chelation therapy by systemic intravenous infusion.

8.2 Limitations of this project and recommendation for future work

1) Mechanisms of how calcified matrix trigger osteogenic response in VSMCs

It is unclear as to how calcified matrix triggers VSMCs transformation to

osteoblast like cells and further cellular pathway studies are needed to test this. Calcium

and phosphorus levels in the media were similar in controls and calcified matrix groups

(data not shown), thus osteoblast-like transformation seen on our studies was not due to

higher amounts of free ions as shown by others[71, 208, 215]. We speculate that VSMCs

synthesize small quantities of bone proteins (they come from same mesenchymal origin

of osteoblasts). These proteins in healthy state do not accumulate in the vessel media;

however, calcified elastin matrix can bind to these synthesized proteins[224, 225] and

increase local concentration causing VSMCs to turn to osteoblast-like cells. We further

speculate that local increase in bone morphogenetic proteins (BMPs) can activate BMP

receptor pathway in SMCs thus causing osteogenic dedifferentiation.

2) Long term therapeutic effects and side effects of systemic EDTA chelation

therapy

EDTA chelation therapy in this work was performed for a period of only two

weeks. Thus, we do not know if long-term EDTA treatment will have therapeutic effects

Page 195: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

177

to reverse vascular calcification or more severe systemic side effects. Others have shown

that long-term EDTA chelation therapy does cause systemic side effects. Secondly, we

did not investigate possible side effects besides serum calcium, urine calcium, and bone

loss. More kidney functional evaluation might be necessary such as serum creatinine

level.

3) Targeted EDTA chelation therapy to reverse calcification using elastin

antibody coated and EDTA loaded albumin NPs

We have clearly shown in chapter 7 that systemic delivery of EDTA loaded NPs

remove calcium from arteries in rats. A lot more work needs to be done to guarantee

limited variation for nanoparticle size, loading efficiency and release between batch-to-

batch preparations for EDTA loaded EL-NPs.

We also do not know whether calcification would return after nanoparticles

deliver all EDTA and resorb calcification. A long term animal study is needed to confirm

that once calcium is removed, it does not return. We also did not test cellular

dedifferentiation in vivo. It would be interesting to see if VSMCs surrounding calcified

matrix show osteoblast-like behavior and whether they return to their native smooth

muscle cell behavior after removal of calcium in vivo similar to what was observed in

vitro in cell cultures.

Only one animal model was used in our current study, where MAC was induced

by chemical treatment. We need to test it different animal models of vascular

calcification, such as animal model for systemic vascular calcification (warfarin +

Page 196: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

178

vitamin K injections in rats) [96], and atherosclerosis (fat-fed apoE-/- mice) [255] by

collaboration with other labs.

In our NPs’ bio-distribution study, we only examined it after 24 hours of

injection of NPs, longer time points, 3 or 7 days, might be necessary to see how long the

NPs could stay. Also, more toxicity tests need to be done on animals such as renal

toxicity, liver toxicity, and trace metal deficiency. Careful risk-benefit analysis needs to

done before use in individual human patients.

Page 197: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

179

CHAPTER 9: REFERENCES

1. Valderrama, A.L., et al., Million Hearts: Strategies to Reduce the Prevalence of

Leading Cardiovascular Disease Risk Factors- United States, 2011. Morbidity

and Mortality Weekly Report, 2011. 60(36).

2. Johnson, R., J. Leopold, and J. Loscalzo, Vascular Calcification: Pathobiological

Mechanisms and Clinical Implications (vol 99, pg 1044, 2006). Circulation

Research, 2009. 105(6): p. E8-E8.

3. Proudfoot, D. and C.M. Shanahan, Biology of Calcification in Vascular Cells:

Intima versus Media. Herz, 2001. 26(4): p. 245-251.

4. MacIsaac, A.I., et al., High speed rotational atherectomy: outcome in calcified

and noncalcified coronary artery lesions. Journal of the American College of

Cardiology, 1995. 26(3): p. 731-736.

5. Coselli, J.S., L.D. Conklin, and S.A. LeMaire, Thoracoabdominal aortic

aneurysm repair: review and update of current strategies. The Annals of thoracic

surgery, 2002. 74(5): p. S1881-S1884.

6. Kapustin, A. and C.M. Shanahan, Targeting vascular calcification: softening-up a

hard target. Current opinion in pharmacology, 2009. 9(2): p. 84-89.

7. Wu, M., C. Rementer, and C.M. Giachelli, Vascular calcification: an update on

mechanisms and challenges in treatment. Calcified tissue international, 2013.

93(4): p. 365-373.

8. Jackson, G., Chelation therapy is TACT-less. International Journal of Clinical

Practice, 2008. 62(12): p. 1821-1822.

9. Atwood, K.C., et al., Why the NIH Trial to Assess Chelation Therapy (TACT)

should be abandoned. Medscape J Med, 2008. 10(5): p. 115.

10. Fraker, T.D. and et al., 2007 Chronic Angina Focused Update of the ACC/AHA

2002 Guidelines for the Management of Patients With Chronic Stable Angina.

Circulation, 2007. 116(23): p. 2762-2772.

11. Kaman, R.L., et al., Effect of EDTA chelation therapy on aortic calcium in rabbits

on atherogenic diets: quantitative and histochemical studies. J Adv Med, 1990.

3(1): p. 13.

12. O’Brien, K.D., Pathogenesis of Calcific Aortic Valve Disease A Disease Process

Comes of Age (and a Good Deal More). Arteriosclerosis, thrombosis, and

vascular biology, 2006. 26(8): p. 1721-1728.

13. Cavallotti, C., et al., Experimental calcification of the aorta in rabbits: effects of

chelating agents and glucagon. J Lab Anim Sci, 2004. 31: p. 215-219.

14. Guldager, B., et al., Effects of intravenous EDTA treatment on serum parathyroid

hormone (1-84) and biochemical markers of bone turnover. Danish medical

bulletin, 1993. 40(5): p. 627-630.

15. Van Rij, A.M., et al., Chelation therapy for intermittent claudication. A double-

blind, randomized, controlled trial. Circulation, 1994. 90(3): p. 1194-1199.

16. Knudtson, M.L., et al., Chelation therapy for ischemic heart disease: a

randomized controlled trial. JAMA, 2002. 287(4): p. 481-486.

Page 198: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

180

17. Lamas, G.A., et al., Effect of disodium EDTA chelation regimen on

cardiovascular events in patients with previous myocardial infarction: the TACT

randomized trial. JAMA, 2013. 309(12): p. 1241-1250.

18. Holland, J.F., E. Danielson, and A. Sahagian-Edwards, Use of ethylene diamine

tetra acetic acid in hypercalcemic patients. Experimental Biology and Medicine,

1953. 84(2): p. 359-364.

19. Schwartz, S.L., et al., Studies of the nephrotoxicity of ethylenediaminetetraacetic

acid. Biochemical pharmacology, 1966. 15(3): p. 377-389.

20. Doolan, P.D., et al., An evaluation of the nephrotoxicity of

ethylenediaminetetraacetate and diethylenetriaminepentaacetate in the rat.

Toxicology and applied pharmacology, 1967. 10(3): p. 481-500.

21. SCHWARTZ, S.L., C.B. JOHNSON, and P.D. DOOLAN, Study of the

Mechanism of Renal Vacuologenesis Induced in the Rat by

Ethylenediaminetetraacetate Comparison of the Cellular Activities of Calcium

and Chromium Chelates. Molecular pharmacology, 1970. 6(1): p. 54-60.

22. McDonagh, E., C. Rudolph, and E. Cheraskin, The effect of EDTA chelation

therapy plus supportive multivitamin-trace mineral supplementation upon renal

function: A study in serum creatinine. J Holistic Med, 1982. 4(2): p. 146-151.

23. Oliver, L., R. Mehta, and H. Sarles, Acute renal failure following administration

of ethylenediamine-tetraacetic acid (EDTA). Texas medicine, 1984. 80(2): p. 40.

24. Spencer, H., et al., Studies on the effect of ethylenediaminetetraacetic acid in

hypercalcemia. J Lab Clin Med, 1956. 47(1): p. 29-41.

25. Meltzer, L.E., J. Kitchell, and F. PALMON JR, The long term use, side effects,

and toxicity of disodium ethylenediamine tetraacetic acid (EDTA). The American

journal of the medical sciences, 1961. 242(2): p. 11-17.

26. Rudolph, C., E. McDonagh, and D. Wussow, The effect of intravenous disodium

ethylenediaminetetraacetic acid (EDTA) upon bone density levels. Journal of

Advancement in Medicine, 1988. 1(2): p. 79-85.

27. Basalyga, D.M., et al., Elastin Degradation and Calcification in an Abdominal

Aorta Injury Model Role of Matrix Metalloproteinases. Circulation, 2004.

110(22): p. 3480-3487.

28. Lei, Y., et al., Efficacy of Reversal of Aortic Calcification by Chelating Agents.

Calcified tissue international, 2013. 93(5): p. 426-435.

29. Singh, R. and J.W. Lillard Jr, Nanoparticle-based targeted drug delivery.

Experimental and Molecular Pathology, 2009. 86(3): p. 215-223.

30. Sinha, A., et al., Nanoparticle targeting to diseased vasculature for imaging and

therapy. Nanomedicine: Nanotechnology, Biology and Medicine, 2014.

31. http://www.phschool.com/. Available from:

http://www.phschool.com/science/biology_place/biocoach/cardio2/structure.html.

32. Rosenbloom, J., W. Abrams, and R. Mecham, Extracellular matrix 4: the elastic

fiber. The FASEB Journal, 1993. 7(13): p. 1208-1218.

33. http://histologistics.com/. Available from:

http://histologistics.com/2013/07/14/elastic-fiber-pictures/.

Page 199: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

181

34. http://helpfromthedoctor.com/. Available from:

http://helpfromthedoctor.com/blog/2010/07/27/what-is-a-protein/.

35. http://www.visualphotos.com/. Available from:

http://www.visualphotos.com/image/1x9082695/elastin_and_microfibrils.

36. Isenburg, J.C., D.T. Simionescu, and N.R. Vyavahare, Elastin stabilization in

cardiovascular implants: improved resistance to enzymatic degradation by

treatment with tannic acid. Biomaterials, 2004. 25(16): p. 3293-3302.

37. Ramamurthi, A. and I. Vesely, Evaluation of the matrix-synthesis potential of

crosslinked hyaluronan gels for tissue engineering of aortic heart valves.

Biomaterials, 2005. 26(9): p. 999-1010.

38. Sinha, S., et al., Expression of latent TGF-beta binding proteins and association

with TGF-beta1 and fibrillin-1 following arterial injury. Cardiovascular Research,

2002. 53(4): p. 971-983.

39. Murie, J.A., Vascular surgery–5th edition, RB Rutherford (ed.), 285× 222 mm.

Pp. 2266. Illustrated. 1999. Philadelphia, Pennsylvania: WB Saunders.£ 199.00.

2000, Wiley Online Library.

40. http://www.cphospital.org/. Available from: http://www.cphospital.org/adam/In-

Depth%20Reports/10/000249.php.

41. Groyer, E., et al., [Immunological aspects of atherosclerosis]. Presse medicale

(Paris, France: 1983), 2006. 35(3 Pt 2): p. 475-486.

42. Black, S., I. Kushner, and D. Samols, C-reactive protein. Journal of Biological

Chemistry, 2004. 279(47): p. 48487-48490.

43. van Oostrom, A.J., J.P. van Wijk, and M.C. Cabezas, Lipaemia, inflammation and

atherosclerosis: novel opportunities in the understanding and treatment of

atherosclerosis. Drugs, 2004. 64(2): p. 19-41.

44. De Caterina, R., J.K. Liao, and P. Libby, Fatty acid modulation of endothelial

activation. The American journal of clinical nutrition, 2000. 71(1): p. 213S-223S.

45. Cummings, P.M., K. Giddens, and B.A. Nassar, Oral glucose loading acutely

attenuates endothelium-dependent vasodilation in healthy adults without

diabetes: an effect prevented by vitamins C and E. Journal of the American

College of Cardiology, 2000. 36(7): p. 2185-2191.

46. Carluccio, M.A., et al., Olive oil and red wine antioxidant polyphenols inhibit

endothelial activation antiatherogenic properties of mediterranean diet

phytochemicals. Arteriosclerosis, thrombosis, and vascular biology, 2003. 23(4):

p. 622-629.

47. D'Acquisto, F., M.J. May, and S. Ghosh, Inhibition of nuclear factor kappa B

(NF-B). Molecular interventions, 2002. 2(1): p. 22.

48. Kopp, E. and S. Ghosh, Inhibition of NF-kappa B by sodium salicylate and

aspirin. Science, 1994. 265(5174): p. 956-959.

49. Grip, O., S. Janciauskiene, and S. Lindgren, Atorvastatin activates PPAR-γ and

attenuates the inflammatory response in human monocytes. Inflammation

Research, 2002. 51(2): p. 58-62.

Page 200: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

182

50. Frick, M.H., et al., Prevention of the angiographic progression of coronary and

vein-graft atherosclerosis by gemfibrozil after coronary bypass surgery in men

with low levels of HDL cholesterol. Circulation, 1997. 96(7): p. 2137-2143.

51. Nordmann, A.J., et al., Balloon angioplasty or medical therapy for hypertensive

patients with atherosclerotic renal artery stenosis? A meta-analysis of

randomized controlled trials. The American journal of medicine, 2003. 114(1): p.

44-50.

52. Lloyd-Jones, D., et al., Heart disease and stroke statistics—2010 update A report

from the American Heart Association. Circulation, 2010. 121(7): p. e46-e215.

53. http://www.wecareindia.com/. Available from:

http://www.wecareindia.com/general-surgery/aortic-aneurysm-surgery.html.

54. Steinmetz, E.F., et al., Treatment with simvastatin suppresses the development of

experimental abdominal aortic aneurysms in normal and hypercholesterolemic

mice. Annals of surgery, 2005. 241(1): p. 92.

55. Brophy, C.M., J.E. Tilson, and M.D. Tilson, Propranolol stimulates the

crosslinking of matrix components in skin from the aneurysm-prone blotchy

mouse. Journal of Surgical Research, 1989. 46(4): p. 330-332.

56. Sho, E., et al., Continuous periaortic infusion improves doxycycline efficacy in

experimental aortic aneurysms. Journal of vascular surgery, 2004. 39(6): p. 1312-

1321.

57. Dai, J., et al., Effect of blocking platelet activation with AZD6140 on development

of abdominal aortic aneurysm in a rat aneurysmal model. Journal of vascular

surgery, 2009. 49(3): p. 719-727.

58. Gavrila, D., et al., Vitamin E inhibits abdominal aortic aneurysm formation in

angiotensin II–infused apolipoprotein E–deficient mice. Arteriosclerosis,

thrombosis, and vascular biology, 2005. 25(8): p. 1671-1677.

59. Walton, L.J., et al., Inhibition of Prostaglandin E2 Synthesis in Abdominal Aortic

Aneurysms Implications for Smooth Muscle Cell Viability, Inflammatory

Processes, and the Expansion of Abdominal Aortic Aneurysms. Circulation, 1999.

100(1): p. 48-54.

60. Go, A.S., et al., Executive Summary: Heart Disease and Stroke Statistics—2013

Update A Report From the American Heart Association. Circulation, 2013.

127(1): p. 143-152.

61. Lau, W.L. and J.H. Ix. Clinical Detection, Risk Factors, and Cardiovascular

Consequences of Medial Arterial Calcification: A Pattern of Vascular Injury

Associated With Aberrant Mineral Metabolism. in Seminars in nephrology. 2013:

Elsevier.

62. Everhart, J., et al., Medial arterial calcification and its association with mortality

and complications of diabetes. Diabetologia, 1988. 31(1): p. 16-23.

63. Giachelli, C.M., Vascular calcification mechanisms. Journal of the American

Society of Nephrology, 2004. 15(12): p. 2959-2964.

64. Luo, G., et al., Spontaneous calcification of arteries and cartilage in mice lacking

matrix GLA protein. 1997.

Page 201: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

183

65. Rutsch, F., et al., Mutations in ENPP1 are associated with'idiopathic'infantile

arterial calcification. Nature genetics, 2003. 34(4): p. 379-381.

66. Giachelli, C.M., et al., Osteopontin is elevated during neointima formation in rat

arteries and is a novel component of human atherosclerotic plaques. Journal of

Clinical Investigation, 1993. 92(4): p. 1686.

67. Levy, R.J., et al., Biologic determinants of dystrophic calcification and

osteocalcin deposition in glutaraldehyde-preserved porcine aortic valve leaflets

implanted subcutaneously in rats. The American journal of pathology, 1983.

113(2): p. 143.

68. Boström, K., et al., Bone morphogenetic protein expression in human

atherosclerotic lesions. Journal of Clinical Investigation, 1993. 91(4): p. 1800.

69. Price, P.A., J.M. Caputo, and M.K. Williamson, Bone Origin of the Serum

Complex of Calcium, Phosphate, Fetuin, and Matrix Gla Protein: Biochemical

Evidence for the Cancellous Bone‐Remodeling Compartment. Journal of Bone

and Mineral Research, 2002. 17(7): p. 1171-1179.

70. Tanimura, A., D.H. McGregor, and H.C. Anderson, Matrix vesicles in

atherosclerotic calcification. Experimental Biology and Medicine, 1983. 172(2):

p. 173-177.

71. Moe, S.M. and N.X. Chen, Mechanisms of Vascular Calcification in Chronic

Kidney Disease. Journal of the American Society of Nephrology, 2008. 19(2): p.

213-216.

72. Parhami, F., et al., Lipid oxidation products have opposite effects on calcifying

vascular cell and bone cell differentiation A possible explanation for the paradox

of arterial calcification in osteoporotic patients. Arteriosclerosis, thrombosis, and

vascular biology, 1997. 17(4): p. 680-687.

73. Tintut, Y., et al., Tumor necrosis factor-α promotes in vitro calcification of

vascular cells via the cAMP pathway. Circulation, 2000. 102(21): p. 2636-2642.

74. Abedin, M., Y. Tintut, and L.L. Demer, Vascular calcification mechanisms and

clinical ramifications. Arteriosclerosis, thrombosis, and vascular biology, 2004.

24(7): p. 1161-1170.

75. Nelson, R.G., et al., Lower-extremity amputations in NIDDM: 12-yr follow-up

study in Pima Indians. Diabetes care, 1988. 11(1): p. 8-16.

76. Lehto, S., et al., Medial Artery Calcification: A Neglected Harbinger of

Cardiovascular Complications in Non–Insulin-Dependent Diabetes Mellitus.

Arteriosclerosis, thrombosis, and vascular biology, 1996. 16(8): p. 978-983.

77. Top, C., et al., Mönckeberg's Sclerosis: An Unusual Presentation A Case Report.

Angiology, 2002. 53(4): p. 483-486.

78. Bas, A., et al., Reversibility of Calcitriol‐Induced Medial Artery Calcification in

Rats With Intact Renal Function. Journal of Bone and Mineral Research, 2006.

21(3): p. 484-490.

79. Al-Aly, Z., et al., Aortic Msx2-Wnt Calcification Cascade Is Regulated by TNF-

α–Dependent Signals in Diabetic Ldlr−/− Mice. Arteriosclerosis, thrombosis, and

vascular biology, 2007. 27(12): p. 2589-2596.

Page 202: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

184

80. Rajamannan, N.M., et al., Human aortic valve calcification is associated with an

osteoblast phenotype. Circulation, 2003. 107(17): p. 2181-2184.

81. Katz, R., et al., Features of the metabolic syndrome and diabetes mellitus as

predictors of aortic valve calcification in the Multi-Ethnic Study of

Atherosclerosis. Circulation, 2006. 113(17): p. 2113-2119.

82. Rajamannan, N.M., R.O. Bonow, and S.H. Rahimtoola, Calcific aortic stenosis:

an update. Nature clinical practice Cardiovascular medicine, 2007. 4(5): p. 254-

262.

83. Coates, T., et al., Cutaneous necrosis from calcific uremic arteriolopathy.

American journal of kidney diseases, 1998. 32(3): p. 384-391.

84. Janigan, D.T., et al., Calcified subcutaneous arterioles with infarcts of the

subcutis and skin (“calciphylaxis”) in chronic renal failure. American journal of

kidney diseases, 2000. 35(4): p. 588-597.

85. Wilmer, W.A. and C.M. Magro. Calciphylaxis: emerging concepts in prevention,

diagnosis, and treatment. in Seminars in dialysis. 2002: Wiley Online Library.

86. Griethe, W., et al., Bone morphogenic protein-4 expression in vascular lesions of

calciphylaxis. Journal of nephrology, 2002. 16(5): p. 728-732.

87. Demer, L.L. and Y. Tintut, Vascular calcification pathobiology of a multifaceted

disease. Circulation, 2008. 117(22): p. 2938-2948.

88. Towler, D.A., Vascular calcification: a perspective on an imminent disease

epidemic. IBMS BoneKEy, 2008. 5(2): p. 41-58.

89. Ikonomidis, J.S., et al., A murine model of thoracic aortic aneurysms. Journal of

Surgical Research, 2003. 115(1): p. 157-163.

90. Gertz, S.D., A. Kurgan, and D. Eisenberg, Aneurysm of the rabbit common

carotid artery induced by periarterial application of calcium chloride in vivo.

Journal of Clinical Investigation, 1988. 81(3): p. 649.

91. Lai, C.-H., et al., Recombinant human thrombomodulin suppresses experimental

abdominal aortic aneurysms induced by calcium chloride in mice. Annals of

surgery, 2013. 258(6): p. 1103-1110.

92. Marinov, G.R., et al., Can the infusion of elastase in the abdominal aorta of the

Yucatan miniature swine consistently produce experimental aneurysms?

Investigative Surgery, 1997. 10(3): p. 129-150.

93. Carsten III, C.G., et al., Elastase is not sufficient to induce experimental

abdominal aortic aneurysms. Journal of vascular surgery, 2001. 33(6): p. 1255-

1262.

94. Ding, Y.H., et al., Modified technique to create morphologically reproducible

elastase-induced aneurysms in rabbits. Neuroradiology, 2006. 48(8): p. 528-532.

95. Gadeau, A.-P., et al., Time course of osteopontin, osteocalcin, and osteonectin

accumulation and calcification after acute vessel wall injury. Journal of

Histochemistry & Cytochemistry, 2001. 49(1): p. 79-86.

96. Price, P.A., S.A. Faus, and M.K. Williamson, Warfarin causes rapid calcification

of the elastic lamellae in rat arteries and heart valves. Arteriosclerosis,

thrombosis, and vascular biology, 1998. 18(9): p. 1400-1407.

Page 203: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

185

97. Bouvet, C., et al., A new rat model of diabetic macrovascular complication.

Cardiovascular Research, 2007. 73(3): p. 504-511.

98. Boström, K. and L.L. Demer, Regulatory mechanisms in vascular calcification.

Critical reviews in eukaryotic gene expression, 1999. 10(2): p. 151-158.

99. Price, P.A., et al., Osteoprotegerin inhibits artery calcification induced by

warfarin and by vitamin D. Arteriosclerosis, thrombosis, and vascular biology,

2001. 21(10): p. 1610-1616.

100. Zittermann, A. and R. Koerfer, Protective and toxic effects of vitamin D on

vascular calcification: clinical implications. Molecular aspects of medicine, 2008.

29(6): p. 423-432.

101. Niederhoffer, N., et al., Aortic calcification produced by vitamin D3 plus nicotine.

Journal of vascular research, 1997. 34(5): p. 386-398.

102. Pan, C.S., et al., The role of adrenomedullin and its receptor system in

cardiovascular calcification of rat induced by Vitamin D3 plus nicotine. Peptides,

2004. 25(4): p. 601-608.

103. Park, J.-H., et al., Relationship between arterial calcification and bone loss in a

new combined model rat by ovariectomy and vitamin D3 plus nicotine. Calcified

tissue international, 2008. 83(3): p. 192-201.

104. Zhou, Y.-B., et al., Lanthanum Acetate Inhibits Vascular Calcification Induced by

Vitamin D3 Plus Nicotine in Rats. Experimental Biology and Medicine, 2009.

234(8): p. 908-917.

105. Adijiang, A., et al., Indoxyl sulphate promotes aortic calcification with expression

of osteoblast-specific proteins in hypertensive rats. Nephrology Dialysis

Transplantation, 2008. 23(6): p. 1892-1901.

106. Kuro-o, M., et al., Mutation of the mouse klotho gene leads to a syndrome

resembling ageing. nature, 1997. 390(6655): p. 45-51.

107. Schäfer, C., et al., The serum protein α2–Heremans-Schmid glycoprotein/fetuin-A

is a systemically acting inhibitor of ectopic calcification. Journal of Clinical

Investigation, 2003. 112(3): p. 357-366.

108. Razzaque, M.S., et al., Premature aging-like phenotype in fibroblast growth

factor 23 null mice is a vitamin D-mediated process. The FASEB Journal, 2006.

20(6): p. 720-722.

109. Gertz, S.D., A. Kurgan, and D. Eisenberg, Aneurysm of the rabbit common

carotid artery induced by periarterial application of calcium chloride in vivo. J

Clin Invest, 1988. 81(3): p. 649-56.

110. Jono, S., et al., 1,25-Dihydroxyvitamin D3 Increases In Vitro Vascular

Calcification by Modulating Secretion of Endogenous Parathyroid Hormone–

Related Peptide. Circulation, 1998. 98(13): p. 1302-1306.

111. Niwa, T. and M. Ise, Indoxyl sulfate, a circulating uremic toxin, stimulates the

progression of glomerular sclerosis. Journal of Laboratory and Clinical Medicine,

1994. 124(1): p. 96-104.

112. Miyazaki, T., et al., Indoxyl sulfate increases the gene expressions of TGF-β1,

TIMP-1 and pro-αl (I) collagen in uremic rat kidneys. Kidney International

Supplement, 1997(62).

Page 204: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

186

113. Hafner, J., et al., Uremic small-artery disease with medial calcification and

intimal hyperplasia (so-called calciphylaxis): a complication of chronic renal

failure and benefit from parathyroidectomy. Journal of the American Academy of

Dermatology, 1995. 33(6): p. 954-962.

114. Asmus, H.-G., et al., Two year comparison of sevelamer and calcium carbonate

effects on cardiovascular calcification and bone density. Nephrology Dialysis

Transplantation, 2005. 20(8): p. 1653-1661.

115. Russell, R.G.G., et al., Bisphosphonates. Annals of the New York Academy of

Sciences, 2007. 1117(1): p. 209-257.

116. O'Neill, W.C., Treatment of vascular calcification. Kidney international, 2008.

74(11): p. 1376-1378.

117. O'Neill, W.C. and K.A. Lomashvili, Recent progress in the treatment of vascular

calcification. Kidney international, 2010. 78(12): p. 1232-1239.

118. Raggi, P., et al., The ADVANCE study: a randomized study to evaluate the effects

of cinacalcet plus low-dose vitamin D on vascular calcification in patients on

hemodialysis. Nephrology Dialysis Transplantation, 2011. 26(4): p. 1327-1339.

119. Pasch, A., et al., Sodium thiosulfate prevents vascular calcifications in uremic

rats. Kidney international, 2008. 74(11): p. 1444-1453.

120. Adirekkiat, S., et al., Sodium thiosulfate delays the progression of coronary artery

calcification in haemodialysis patients. Nephrology Dialysis Transplantation,

2010. 25(6): p. 1923-1929.

121. Mathews, S.J., et al., Effects of sodium thiosulfate on vascular calcification in

end-stage renal disease: a pilot study of feasibility, safety and efficacy. American

journal of nephrology, 2011. 33(2): p. 131-138.

122. Ong, S. and I. Coulson, Diagnosis and treatment of calciphylaxis. Skinmed, 2012.

10(3): p. 166-170.

123. Callister, T.Q., et al., Effect of HMG-CoA reductase inhibitors on coronary artery

disease as assessed by electron-beam computed tomography. New England

Journal of Medicine, 1998. 339(27): p. 1972-1978.

124. Novaro, G.M., et al., Effect of hydroxymethylglutaryl coenzyme a reductase

inhibitors on the progression of calcific aortic stenosis. Circulation, 2001.

104(18): p. 2205-2209.

125. Wanner, C., et al., Atorvastatin in patients with type 2 diabetes mellitus

undergoing hemodialysis. New England Journal of Medicine, 2005. 353(3): p.

238-248.

126. Mach, F., Statins as immunomodulatory agents. Circulation, 2004. 109(21 suppl

1): p. II-15-II-17.

127. Greenwood, J., L. Steinman, and S.S. Zamvil, Statin therapy and autoimmune

disease: from protein prenylation to immunomodulation. Nature Reviews

Immunology, 2006. 6(5): p. 358-370.

128. Spronk, H., et al., Tissue-specific utilization of menaquinone-4 results in the

prevention of arterial calcification in warfarin-treated rats. Journal of vascular

research, 2004. 40(6): p. 531-537.

Page 205: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

187

129. Holden, R.M., et al., Vitamins K and D status in stages 3–5 chronic kidney

disease. Clinical Journal of the American Society of Nephrology, 2010. 5(4): p.

590-597.

130. Schlieper, G., et al., Circulating nonphosphorylated carboxylated matrix gla

protein predicts survival in ESRD. Journal of the American Society of

Nephrology, 2011. 22(2): p. 387-395.

131. Westenfeld, R., et al., Effect of Vitamin K< sub> 2</sub> Supplementation on

Functional Vitamin K Deficiency in Hemodialysis Patients: A Randomized Trial.

American journal of kidney diseases, 2012. 59(2): p. 186-195.

132. Rajachar, R.M., et al., Role of carbonic anhydrase II in ectopic calcification.

Cardiovascular Pathology, 2009. 18(2): p. 77-82.

133. Simpson, C.L., et al., Toward cell therapy for vascular calcification: osteoclast-

mediated demineralization of calcified elastin. Cardiovascular Pathology, 2007.

16(1): p. 29-37.

134. Bolick, L.E. and D.H. Blankenhorn, A quantitative study of coronary arterial

calcification. The American journal of pathology, 1961. 39(5): p. 511.

135. Maxwell, G., R. Elliott, and E. Robertson, The effect of Na< sub> 3</sub>

EDTA-induced hypocalcemia upon the general and coronary hemodynamics of

the intact animal. American heart journal, 1963. 66(1): p. 82-87.

136. Lufkin, E.G., R. Kumar, and H. Heath III, Hyperphosphatemic Tumoral

Calcinosis: Effects of Phosphate Depletion on Vitamin D Metabolism, and of

Acute Hypocalcemia on Parathyroid Hormone Secretion and Action*. The

Journal of Clinical Endocrinology & Metabolism, 1983. 56(6): p. 1319-1322.

137. Olszewer, E., F.C. Sabbag, and J.P. Carter, A pilot double-blind study of sodium-

magnesium EDTA in peripheral vascular disease. Journal of the National Medical

Association, 1990. 82(3): p. 173.

138. Cranton, E.M., Bypassing Bypass Surgery: A Non-surgical Treatment for

Reversing Arteriosclerosis, Improving Blocked Circulation, and Slowing the

Aging Process. 2001: Hampton Roads Publishing.

139. Maniscalco, B.S. and K.A. Taylor, Calcification in coronary artery disease can

be reversed by EDTA–tetracycline long-term chemotherapy. Pathophysiology,

2004. 11(2): p. 95-101.

140. Silay, M.S. and C. Miroglu, The risk of urolithiasis recurrence may be reduced

with anti-nanobacterial therapy. Medical hypotheses, 2007. 68(6): p. 1348-1350.

141. Lamas, G.A. and S.J. Hussein, EDTA chelation therapy meets evidence-based

medicine. Complementary Therapies in Clinical Practice, 2006. 12(3): p. 213-215.

142. Clarke, N.E., C.N. Clarke, and R.E. Mosher, Treatment of angina pectoris with

disodium ethylene diamine tetraacetic acid. The American journal of the medical

sciences, 1956. 232(6): p. 654-666.

143. Robert Jr, C., Agent affecting calcification, Calcium, parathyroid hormone,

calcitonin, vitamin D and other compound. Goodman Gillman’s the

pharmacological basis of therapeutics. 7th ed. New York: Macmillan Publishing

Co, 1980: p. 1496-522.

Page 206: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

188

144. Sucu, N., et al., Two stage EDTA anti-calcification method for bioprosthetic heart

valve materials. Medical science monitor: international medical journal of

experimental and clinical research, 2006. 12(6): p. MT33-8.

145. Trevor, A.J., B.G. Katzung, and S.B. Masters, Katzung & Trevor's Pharmacology

Examination and Board Review. 2007: McGraw-Hill Education.

146. Saxena, G. and S. Flora, Lead‐ induced oxidative stress and hematological

alterations and their response to combined administration of calcium disodium

EDTA with a thiol chelator in rats. Journal of biochemical and molecular

toxicology, 2004. 18(4): p. 221-233.

147. Leckie, W. and S. Tompsett, The diagnostic and therapeutic use of edathamil

calcium disodium (EDTA, Versene) in excessive inorganic lead absorption. QJM,

1958. 27(1): p. 65-82.

148. Hayden, M.R., et al., Vascular ossification–calcification in metabolic syndrome,

type 2 diabetes mellitus, chronic kidney disease, and calciphylaxis–calcific

uremic arteriolopathy: the emerging role of sodium thiosulfate. Cardiovascular

diabetology, 2005. 4(1): p. 4.

149. Dickey, D.T., et al., Protection against cisplatin-induced toxicities by N-

acetylcysteine and sodium thiosulfate as assessed at the molecular, cellular, and

in vivo levels. Journal of Pharmacology and Experimental Therapeutics, 2005.

314(3): p. 1052-1058.

150. Pouyatos, B., et al., Oxidative stress pathways in the potentiation of noise-induced

hearing loss by acrylonitrile. Hearing research, 2007. 224(1): p. 61-74.

151. Hayden, M.R., et al., Calciphylaxis: calcific uremic arteriolopathy and the

emerging role of sodium thiosulfate. International urology and nephrology, 2008.

40(2): p. 443-451.

152. Caravan, P., et al., Gadolinium (III) chelates as MRI contrast agents: structure,

dynamics, and applications. Chemical reviews, 1999. 99(9): p. 2293-2352.

153. Brown, M.A., A. Paulenova, and A.V. Gelis, Aqueous complexation of thorium

(IV), uranium (IV), neptunium (IV), plutonium (III/IV), and cerium (III/IV) with

DTPA. Inorganic chemistry, 2012. 51(14): p. 7741-7748.

154. Goldman, M. and J.C. Dacre, Lewisite: its chemistry, toxicology, and biological

effects, in Reviews of environmental contamination and toxicology. 1989,

Springer. p. 75-115.

155. Archbold, G., R.M. McGuckin, and N. Campbell, Dimercaptosuccinic acid

loading test for assessing mercury burden in healthy individuals. Annals of

clinical biochemistry, 2004. 41(3): p. 233-236.

156. Aposhian, H.V. and M.M. Aposhian, Meso-2, 3-dimercaptosuccinic acid:

chemical, pharmacological and toxicological properties of an orally effective

metal chelating agent. Annual review of pharmacology and toxicology, 1990.

30(1): p. 279-306.

157. Gonzalez-Ramirez, D., et al., DMPS (2, 3-dimercaptopropane-1-sulfonate,

dimaval) decreases the body burden of mercury in humans exposed to mercurous

chloride. Journal of Pharmacology and Experimental Therapeutics, 1998. 287(1):

p. 8-12.

Page 207: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

189

158. Peisach, J. and W. Blumberg, A mechanism for the action of penicillamine in the

treatment of Wilson’s disease. Molecular pharmacology, 1969. 5(2): p. 200-209.

159. Moeschlin, S. and U. Schnider, Treatment of primary and secondary

haemochromatosis and acute iron poisoning with a new, potent iron-eliminating

agent (desferrioxamine B), in Iron Metabolism. 1964, Springer. p. 525-550.

160. Westlin, W.F., Deferoxamine in the Treatment of Acute Iron Poisoning Clinical

Experiences with 172 Children. Clinical pediatrics, 1966. 5(9): p. 531-535.

161. Halstead, B.W., The scientific basis of EDTA chelation therapy. 1979: Golden

Quill Publishers.

162. Rozema, T.C., The protocol for the safe and effective administration of EDTA and

other chelating agents for vascular disease, degenerative disease, and metal

toxicity. Journal of Advancement in Medicine, 1997. 10(1): p. 5-100.

163. Singla, A. and C.H. Lee, Inhibition of CEM calcification by the sequential

pretreatment with ethanol and EDTA. Journal of Biomedical Materials Research

Part A, 2003. 64(4): p. 706-713.

164. Sucu, N., et al., The effect of ethylenediaminetetraacetic acid on calcific

degeneration in bovine pericardium. Heart and vessels, 2004. 19(2): p. 89-93.

165. O'Brien, S., et al., Chelation therapy in the JCR: LA-cp rat: experimental

assessment of a putative antiatherosclerotic treatment. Clinical and investigative

medicine. Medecine clinique et experimentale, 2000. 23(2): p. 124-131.

166. O’Neill, W.C. and K.I. Hardcastle, The chemistry of thiosulfate and vascular

calcification. Nephrology Dialysis Transplantation, 2012. 27(2): p. 521-526.

167. Clarke Sr, N.E., Atherosclerosis, occlusive vascular disease and EDTA. The

American journal of cardiology, 1960. 6(2): p. 233-236.

168. Rudolph, C., E. McDonagh, and R. Barber, A non-surgical approach to

obstructive carotid stenosis using EDTA chelation. J Adv Med, 1991. 4(3): p.

157.

169. Hancke, C. and K. Flytlie, Benefits of EDTA chelation therapy in arteriosclerosis:

a retrospective study of 470 patients. Journal of Advancement in Medicine, 1993.

6(3): p. 161-171.

170. Shoskes, D.A., K.D. Thomas, and E. Gomez, ANTI-NANOBACTERIAL

THERAPY FOR MEN WITH CHRONIC PROSTATITIS/CHRONIC PELVIC

PAIN SYNDROME AND PROSTATIC STONES: PRELIMINARY EXPERIENCE.

The Journal of Urology, 2005. 173(2): p. 474-477.

171. Cacchio, A., et al., Effectiveness of treatment of calcific tendinitis of the shoulder

by disodium EDTA. Arthritis Care & Research, 2009. 61(1): p. 84-91.

172. Cranton, E.M., A textbook on EDTA chelation therapy. 2001: Hampton Roads

Publishing.

173. Soffer, A., Chelation therapy for cardiovascular disease. Chelation Therapy,

Springfield, Illinois, Charles C. Thomas, 1964: p. 15-33.

174. Fraker, T.D. and S.D. Fihn, 2007 Chronic Angina Focused Update of the

ACC/AHA 2002 Guidelines for the Management of Patients With Chronic Stable

AnginaA Report of the American College of Cardiology/American Heart

Association Task Force on Practice Guidelines Writing Group to Develop the

Page 208: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

190

Focused Update of the 2002 Guidelines for the Management of Patients With

Chronic Stable Angina. Journal of the American College of Cardiology, 2007.

50(23): p. 2264-2274.

175. Jackson, G., Chelation therapy is TACT‐less. International Journal of Clinical

Practice, 2008. 62(12): p. 1821-1822.

176. Atwood IV, K.C., et al., Why the NIH Trial to Assess Chelation Therapy (TACT)

should be abandoned. The Medscape Journal of Medicine, 2008. 10(5): p. 115.

177. Jo̸rgensen, B., et al., Femoral artery recanalisation with percutaneous

angioplasty and segmentally enclosed plasminogen activator. The Lancet, 1989.

333(8647): p. 1106-1108.

178. Ikeno, F., et al., Novel percutaneous adventitial drug delivery system for regional

vascular treatment. Catheterization and cardiovascular interventions, 2004. 63(2):

p. 222-230.

179. Villa, A.E., et al., Local delivery of dexamethasone for prevention of neointimal

proliferation in a rat model of balloon angioplasty. Journal of Clinical

Investigation, 1994. 93(3): p. 1243.

180. Yamawaki-Ogata, A., et al., A doxycycline loaded, controlled-release,

biodegradable fiber for the treatment of aortic aneurysms. Biomaterials, 2010.

31(36): p. 9554-9564.

181. Valéro, F., et al., Intramural injection of biodegradable microspheres as a local

drug-delivery system to inhibit neointimal thickening in a rabbit model of balloon

angioplasty. Journal of cardiovascular pharmacology, 1998. 31(4): p. 513-519.

182. Westedt, U., et al., Deposition of nanoparticles in the arterial vessel by porous

balloon catheters: localization by confocal laser scanning microscopy and

transmission electron microscopy. AAPS PharmSci, 2002. 4(4): p. 206-211.

183. Elzoghby, A.O., W.M. Samy, and N.A. Elgindy, Albumin-based nanoparticles as

potential controlled release drug delivery systems. Journal of Controlled Release,

2012. 157(2): p. 168-182.

184. Tantra, R., J. Tompkins, and P. Quincey, Characterisation of the de-

agglomeration effects of bovine serum albumin on nanoparticles in aqueous

suspension. Colloids and Surfaces B: Biointerfaces, 2010. 75(1): p. 275-281.

185. Kratz, F., Albumin as a drug carrier: design of prodrugs, drug conjugates and

nanoparticles. Journal of Controlled Release, 2008. 132(3): p. 171-183.

186. Langer, K., et al., Optimization of the preparation process for human serum

albumin (HSA) nanoparticles. International journal of pharmaceutics, 2003.

257(1): p. 169-180.

187. Patil, G.V., Biopolymer albumin for diagnosis and in drug delivery. Drug

development research, 2003. 58(3): p. 219-247.

188. Yu, S., et al., Nanogels prepared by self‐ assembly of oppositely charged

globular proteins. Biopolymers, 2006. 83(2): p. 148-158.

189. Lee, S.H., et al., Nano spray drying: a novel method for preparing protein

nanoparticles for protein therapy. International journal of pharmaceutics, 2011.

403(1): p. 192-200.

Page 209: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

191

190. Desai, N., Nanoparticle albumin bound (< i> nab</i>) technology: targeting

tumors through the endothelial gp60 receptor and SPARC. Nanomedicine:

Nanotechnology, Biology and Medicine, 2007. 3(4): p. 339.

191. Xu, R., M. Fisher, and R. Juliano, Targeted albumin-based nanoparticles for

delivery of amphipathic drugs. Bioconjugate chemistry, 2011. 22(5): p. 870-878.

192. Merodio, M., et al., Ganciclovir-loaded albumin nanoparticles: characterization

and in vitro release properties. European journal of pharmaceutical sciences,

2001. 12(3): p. 251-259.

193. Segura, S., et al., Gamma interferon loaded onto albumin nanoparticles: in vitro

and in vivo activities against Brucella abortus. Antimicrobial agents and

chemotherapy, 2007. 51(4): p. 1310-1314.

194. Das, S., R. Banerjee, and J. Bellare, Aspirin loaded albumin nanoparticles by

coacervation: implications in drug delivery. Trends Biomater Artif Organs, 2005.

18(2): p. 203-212.

195. Wagner, S., et al., Enhanced drug targeting by attachment of an anti αv integrin

antibody to doxorubicin loaded human serum albumin nanoparticles.

Biomaterials, 2010. 31(8): p. 2388-2398.

196. Löw, K., et al., Targeted human serum albumin nanoparticles for specific uptake

in EGFR-Expressing colon carcinoma cells. Nanomedicine: Nanotechnology,

Biology and Medicine, 2011. 7(4): p. 454-463.

197. Bini, A., et al., Noncollagenous Bone Matrix Proteins, Calcification, and

Thrombosis in Carotid Artery Atherosclerosis. Arteriosclerosis, thrombosis, and

vascular biology, 1999. 19(8): p. 1852-1861.

198. Dhore, C.R., et al., Differential Expression of Bone Matrix Regulatory Proteins in

Human Atherosclerotic Plaques. Arteriosclerosis, thrombosis, and vascular

biology, 2001. 21(12): p. 1998-2003.

199. Basalyga, D.M., et al., Elastin Degradation and Calcification in an Abdominal

Aorta Injury Model: Role of Matrix Metalloproteinases. Circulation, 2004.

110(22): p. 3480-3487.

200. Lee, J.S., et al., Elastin Calcification in the Rat Subdermal Model Is Accompanied

by Up-Regulation of Degradative and Osteogenic Cellular Responses. The

American journal of pathology, 2006. 168(2): p. 490-498.

201. Stegemann, J.P. and R.M. Nerem, Altered response of vascular smooth muscle

cells to exogenous biochemical stimulation in two-and three-dimensional culture.

Experimental cell research, 2003. 283(2): p. 146-155.

202. Mavrilas, D., et al., Development of bioprosthetic heart valve calcification in vitro

and in animal models: morphology and composition. Journal of Crystal Growth,

1999. 205(4): p. 554-562.

203. Simionescu, A., D.T. Simionescu, and N.R. Vyavahare, Osteogenic Responses in

Fibroblasts Activated by Elastin Degradation Products and Transforming Growth

Factor-β1: Role of Myofibroblasts in Vascular Calcification. The American

journal of pathology, 2007. 171(1): p. 116-123.

204. Low, R.B., et al., Age dependence of smooth muscle myosin expression by

cultured rat aortic smooth muscle cells. Differentiation, 1999. 65(3): p. 151-159.

Page 210: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

192

205. Agung, M., et al., Mobilization of bone marrow-derived mesenchymal stem cells

into the injured tissues after intraarticular injection and their contribution to

tissue regeneration. Knee Surgery, Sports Traumatology, Arthroscopy, 2006.

14(12): p. 1307-1314.

206. Zhang, M., et al., IGF-1 regulation of type II collagen and MMP-13 expression in

rat endplate chondrocytes< i> via</i> distinct signaling pathways. Osteoarthritis

and Cartilage, 2009. 17(1): p. 100-106.

207. Livak, K.J. and T.D. Schmittgen, Analysis of Relative Gene Expression Data

Using Real-Time Quantitative PCR and the 2< sup>− ΔΔCT</sup> Method.

methods, 2001. 25(4): p. 402-408.

208. Steitz, S.A., et al., Smooth Muscle Cell Phenotypic Transition Associated With

Calcification: Upregulation of Cbfa1 and Downregulation of Smooth Muscle

Lineage Markers. Circulation Research, 2001. 89(12): p. 1147-1154.

209. Giachelli, C.M., et al., Regulation of Vascular Calcification: Roles of Phosphate

and Osteopontin. Circulation Research, 2005. 96(7): p. 717-722.

210. Franceschi, R.T., The Developmental Control of Osteoblast-Specific Gene

Expression: Role of Specific Transcription Factors and the Extracellular Matrix

Environment. Critical Reviews in Oral Biology & Medicine, 1999. 10(1): p. 40-

57.

211. Ducy, P., et al., Osf2/Cbfa1: A Transcriptional Activator of Osteoblast

Differentiation. Cell, 1997. 89(5): p. 747-754.

212. Simionescu, A., K. Philips, and N. Vyavahare, Elastin-derived peptides and TGF-

β1 induce osteogenic responses in smooth muscle cells. Biochemical and

Biophysical Research Communications, 2005. 334(2): p. 524-532.

213. Oshima, A., et al., A novel mechanism for the regulation of osteoblast

differentiation: Transcription of periostin, a member of the fasciclin I family, is

regulated by the bHLH transcription factor, twist. Journal of Cellular

Biochemistry, 2002. 86(4): p. 792-804.

214. Demer, L.L. and Y. Tintut, Vascular Calcification: Pathobiology of a

Multifaceted Disease. Circulation, 2008. 117(22): p. 2938-2948.

215. Jono, S., et al., Phosphate Regulation of Vascular Smooth Muscle Cell

Calcification. Circulation Research, 2000. 87(7): p. e10-e17.

216. Neven, E., et al., Endochondral bone formation is involved in media calcification

in rats and in men. Kidney international, 2007. 72(5): p. 574-581.

217. Shao, J.-S., et al., Msx2 promotes cardiovascular calcification by activating

paracrine Wnt signals. Journal of Clinical Investigation, 2005. 115(5): p. 1210-

1220.

218. Towler, D.A., Angiogenesis and marrow stromal cell fates: roles in bone

strength. Osteoporosis international, 2003. 14(5): p. 46-53.

219. Chen, N.X., et al., High glucose increases the expression of Cbfa1 and BMP-2

and enhances the calcification of vascular smooth muscle cells. Nephrology

Dialysis Transplantation, 2006. 21(12): p. 3435-3442.

220. Urry, D.W., Neutral Sites for Calcium Ion Binding to Elastin and Collagen: A

Charge Neutralization Theory for Calcification and Its Relationship to

Page 211: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

193

Atherosclerosis. Proceedings of the National Academy of Sciences, 1971. 68(4):

p. 810-814.

221. Price, P.A., et al., The Elastic Lamellae of Devitalized Arteries Calcify When

Incubated in Serum: Evidence for a Serum Calcification Factor. Arteriosclerosis,

thrombosis, and vascular biology, 2006. 26(5): p. 1079-1085.

222. Khavandgar, Z., et al., Elastin haploinsufficiency impedes the progression of

arterial calcification in MGP‐deficient mice. Journal of Bone and Mineral

Research, 2013.

223. Villa-Bellosta, R., A. Millan, and V. Sorribas, Role of calcium-phosphate

deposition in vascular smooth muscle cell calcification. American Journal of

Physiology - Cell Physiology, 2011. 300(1): p. C210-C220.

224. Hoang, Q.Q., et al., Bone recognition mechanism of porcine osteocalcin from

crystal structure. nature, 2003. 425(6961): p. 977-980.

225. Reinholt, F.P., et al., Osteopontin--a possible anchor of osteoclasts to bone.

Proceedings of the National Academy of Sciences, 1990. 87(12): p. 4473-4475.

226. Speer, M.Y. and C.M. Giachelli, Regulation of cardiovascular calcification.

Cardiovascular Pathology, 2004. 13(2): p. 63-70.

227. Lei, Y., et al., Efficacy of Reversal of Aortic Calcification by Chelating Agents.

Calcified Tissue International, 2013: p. 1-10.

228. Liu, Y., et al., Cortistatin attenuates vascular calcification in rats. Regulatory

Peptides, 2010. 159(1-3): p. 35-43.

229. Shanahan, C.M., et al., Medial Localization of Mineralization-Regulating

Proteins in Association With Mönckeberg’s Sclerosis: Evidence for Smooth

Muscle Cell–Mediated Vascular Calcification. Circulation, 1999. 100(21): p.

2168-2176.

230. Lamas Ga, G.C.B.R. and et al., Effect of disodium edta chelation regimen on

cardiovascular events in patients with previous myocardial infarction: The tact

randomized trial. JAMA, 2013. 309(12): p. 1241-1250.

231. Partridge, S. and F. Keeley, Age related and atherosclerotic changes in aortic

elastin, in Arterial Mesenchyme and Arteriosclerosis. 1974, Springer. p. 173-191.

232. Vyavahare, N., et al., Elastin calcification and its prevention with aluminum

chloride pretreatment. The American journal of pathology, 1999. 155(3): p. 973-

982.

233. Labhasetwar, V., C. Song, and R.J. Levy, Nanoparticle drug delivery system for

restenosis. Advanced Drug Delivery Reviews, 1997. 24(1): p. 63-85.

234. Cagnasso, C.E., et al., Development and validation of a method for the

determination of EDTA in non-alcoholic drinks by HPLC. Journal of Food

Composition and Analysis, 2007. 20(3–4): p. 248-251.

235. Basalyga, D.M., et al., Elastin Degradation and Calcification in an Abdominal

Aorta Injury Model. Circulation, 2004. 110(22): p. 3480-3487.

236. Pasch, A., et al., Sodium thiosulfate prevents vascular calcifications in uremic

rats. Kidney Int, 2008. 74(11): p. 1444-1453.

Page 212: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

194

237. Postnov, A.A., et al., Possibilities and limits of X-ray microtomography for in

vivo and ex vivo detection of vascular calcifications. The International Journal of

Cardiovascular Imaging, 2009. 25(6): p. 615-624.

238. C Cavallotti, F.M.T.L., M Artico, Experimental calcification of the aorta in

rabbits: Effects of chelating agents and glucagon. Journal of Laboratory Animal

Science, 2004. 31(4): p. 215-219.

239. Persy, V., et al., High-Resolution X-Ray Microtomography Is a Sensitive Method

to Detect Vascular Calcification in Living Rats With Chronic Renal Failure.

Arteriosclerosis, thrombosis, and vascular biology, 2006. 26(9): p. 2110-2116.

240. Lamas, G.A., et al., Design of the Trial to Assess Chelation Therapy (TACT).

American Heart Journal, 2012. 163(1): p. 7-12.

241. Waters, R.S., et al., EDTA chelation effects on urinary losses of cadmium,

calcium, chromium, cobalt, copper, lead, magnesium, and zinc. Biological trace

element research, 2001. 83(3): p. 207-221.

242. Shroff, R.C. and C.M. Shanahan. Vascular calcification in patients with kidney

disease: The vascular biology of calcification. in Seminars in dialysis. 2007:

Wiley Online Library.

243. Hanken, J. and R. Wassersug, A new double-stain technique reveals the nature of

the hard tissues. Funct. Photog, 1981. 16: p. 22-26.

244. Rosa-Molinar, E., et al., Whole-mount procedures for simultaneous visualization

of nerves, neurons, cartilage and bone. Brain Research Protocols, 1999. 4(2): p.

115-123.

245. Foreman, H. and T. Trujillo, The metabolism of C14 labeled

ethylenediaminetetraacetic acid in human beings. The Journal of laboratory and

clinical medicine, 1954. 43(4): p. 566.

246. Lamas, G.A., et al., Effect of disodium edta chelation regimen on cardiovascular

events in patients with previous myocardial infarction: The tact randomized trial.

JAMA, 2013. 309(12): p. 1241-1250.

247. Hawkins, M.J., P. Soon-Shiong, and N. Desai, Protein nanoparticles as drug

carriers in clinical medicine. Advanced drug delivery reviews, 2008. 60(8): p.

876-885.

248. Wartlick, H., et al., Highly specific HER2-mediated cellular uptake of antibody-

modified nanoparticles in tumour cells. Journal of drug targeting, 2004. 12(7): p.

461-471.

249. Cagnasso, C.E., et al., Development and validation of a method for the

determination of EDTA in non-alcoholic drinks by HPLC. Journal of Food

Composition and Analysis, 2007. 20(3): p. 248-251.

250. Sivaraman, B. and A. Ramamurthi, Multifunctional nanoparticles for doxycycline

delivery towards localized elastic matrix stabilization and regenerative repair.

Acta biomaterialia, 2013. 9(5): p. 6511-6525.

251. Song, C., et al., Arterial uptake of biodegradable nanoparticles for intravascular

local drug delivery: results with an acute dog model. Journal of Controlled

Release, 1998. 54(2): p. 201-211.

Page 213: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

195

252. Cho, E.C., et al., The Effects of Size, Shape, and Surface Functional Group of

Gold Nanostructures on Their Adsorption and Internalization by Cells. Small,

2010. 6(4): p. 517-522.

253. Schrama, D., R.A. Reisfeld, and J.C. Becker, Antibody targeted drugs as cancer

therapeutics. Nature Reviews Drug Discovery, 2006. 5(2): p. 147-159.

254. Kaminski, M.S., et al., Radioimmunotherapy of B-cell lymphoma with [131I] anti-

B1 (anti-CD20) antibody. New England Journal of Medicine, 1993. 329(7): p.

459-465.

255. Lessner, S.M., et al., Atherosclerotic lesions grow through recruitment and

proliferation of circulating monocytes in a murine model. The American journal

of pathology, 2002. 160(6): p. 2145-2155.

Page 214: MECHANISMS AND REVERSAL OF ELASTIN SPECIFIC MEDIAL

196

SUPPLEMENTAL FILES

Supplemental videos online:

1) Micro CT for rat aortas of 2 groups (Saline and HD EDTA).

2) Micro CT rat femoral bone of 2 groups (Saline and HD EDTA).

3) Three points bending test.

4) Micro CT for rat aortas of 5 groups (Saline, HD EDTA, LD EDTA, IgG-

NPs/EDTA, and EL-NPs/EDTA).

5) Micro CT rat femoral bone of 5 groups (Saline, HD EDTA, LD EDTA, IgG-

NPs/EDTA, and EL-NPs/EDTA).