complement regulation on vascular endothelial cells ...€¦ · 2.3.8.2 bioflux (fluxion...

136
Complement Regulation on Vascular Endothelial Cells Insights into the Pathogenesis of Thrombotic Microangiopathy by Damien Noone A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright 2015 by Damien G. Noone

Upload: others

Post on 23-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

Complement Regulation on Vascular Endothelial

Cells – Insights into the Pathogenesis of Thrombotic

Microangiopathy

by

Damien Noone

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Institute of Medical Science

University of Toronto

© Copyright 2015 by Damien G. Noone

Page 2: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

II

Complement Regulation On Vascular Endothelial Cells – Insights

Into The Pathogenesis Of Thrombotic Microangiopathy

Damien Noone

Master of Science

Institute of Medical Science

University of Toronto

2015

Abstract

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic

microangiopathy (TMA) that occurs due to defective regulation of the alternative

complement pathway (AP). We developed a novel model system using Blood

Outgrowth Endothelial Cells (BOEC), whereby the response of these cells to

complement challenge could be examined under static and microfluidic conditions, in

order to study aHUS pathogenesis. Complete blockade of the membrane-anchored,

AP regulator CD46/MCP, associated with disease in patients, was insufficient to

cause an ex vivo TMA phenotype. Increasing „complement challenge‟, mimicking

additional genetic „hits‟ in complement regulation achieved the phenotype. In addition,

using BOEC from patients lacking von Willebrand Factor (vWF), a hemostatic

protein released from activated endothelial cells, we showed that contrary to current

opinion, vWF does not amplify complement, rather regulates it. The paucity of vWF

in the glomerular endothelium could explain the vulnerability of the kidney to loss of

complement control in aHUS.

Page 3: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

III

Acknowledgements

I would like to express my sincerest appreciation and heartfelt gratitude to Dr

Christoph Licht who has been my supervisor, mentor and career advisor. You have

been considerably greater than the sum of those parts. I am indebted to you for your

unflinching support. It will be an absolute honor, to be your colleague at The Hospital

for Sick Children and I hope that you will continue as my mentor for the rest of my

career.

I wish to thank Dr Lisa Robinson for her support, direction, constructive feedback on

how to communicate scientific information, and most especially, for helping me in

my transition from clinician to academic clinician.

I wish to thank Dr Philip Marsden for taking interest in my research and for asking

the tough, probing questions that undoubtedly have made me, and this work, a great

deal stronger.

I would like to especially thank Dr Magdalena Riedl. You have been a tremendous

help and guidance over the past year. Your work ethic is awe-inspiring.

I would also like to thank Dr Fred G Pluthero. Your scientific acumen and insight,

imaging skills, general knowledge and scrabble skills have always enlivened my

laboratory experience.

To Mackenzie Bowman and Dr Paula James in Kingston, Ontario, for your time and

patience in teaching me the technique of blood outgrowth endothelial cell culture.

To Kathy Liszewski and Dr John Atkinson in St Louis Missouri - for the gift of your

time, hospitality and knowledge I am indebted.

To the students of the Licht Laboratory who helped along the way – Lily Lu, Yi

Emma Quan and Steve Balgobin – thank you and good luck in your respective careers.

To Dr Norman Rosenblum, Dr Rulan Parekh and Dr Binita Kamath, members of my

Clinical Fellowship Research Advisory Committee at The Hospital for Sick Children,

for their advice and mentorship.

I owe my deepest gratitude to my wife. Thank you for your encouragement, your

endless sacrifice and love. Thank you for your acceptance of the uncertainties that

came with a medical career, two fellowships in two new countries and one Masters.

I dedicate this thesis to my three daughters, Jasmine, Molly and Annabelle.

I would like to acknowledge the following funding sources: Restracomp and The

Transplant Centre at The Hospital for Sick Children.

Page 4: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

IV

Contributions

Damien Noone (author) solely prepared this thesis. All aspects of this body of work,

including the planning, execution, analysis, and writing of all original research and

publications was performed in whole or in part by the author. The following

contributions by other individuals are formally and inclusively acknowledged:

Dr. Christoph Licht (Primary Supervisor and Thesis Committee Member) –

mentorship; laboratory resources; guidance and assistance in planning, execution, and

analysis of experiments as well as thesis preparation

Dr. Lisa Robinson (Supervisor and Thesis Committee Member) – mentorship;

laboratory resources; guidance and assistance in planning, execution, and analysis of

experiments as well as thesis preparation

Dr. Philip Marsden (Thesis Committee Member) – mentorship; guidance in

interpretation of results as well as thesis preparation

Dr Magdalena Riedl - guidance and assistance in planning, execution, and analysis of

experiments for chapters 2 and 3

Dr Fred G Pluthero - guidance and assistance in planning, execution, and analysis of

experiments for chapters 2 and 3

Dr Walter Kahr - laboratory resources; guidance in interpretation of results

Annie Bang - guidance and assistance in planning, execution, and analysis of

experiments for chapter 2

Lily Lu - assistance in execution, and analysis of experiments for chapter 2

Yi (Emma) Quan - guidance and assistance in planning, execution, and analysis of

experiments for chapter 2

Steve Balgobin - assistance in execution, and analysis of experiments for chapters 2

and 3

Page 5: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

V

Table of Contents

ACKNOWLEDGEMENTS………………………………………………………………….……….III

CONTRIBUTIONS………………………………………………………………………….………..IV

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

LIST OF TABLES…………………………………………………………………………………….IX

LIST OF FIGURES…………………………………………………………….…………………..….X

LIST OF ABBREVIATIONS…………………………………………………………………….…XII

Contents

CHAPTER 1. AN INTRODUCTION TO THROMBOTIC

MICROANGIOPATHY .............................................................................................. 1

1.1 Introduction ............................................................................................................ 3

1.2 Thrombotic microangiopathy ............................................................................... 4

1.2.1 Introduction to Thrombotic Microangiopathy ........................................................ 4

1.2.2 The Spectrum of Thrombotic Microangiopathy ..................................................... 4

1.2.3 Thrombotic Thrombocytopenic Purpura, VWF and ADAMTS13 ....................... 6

1.2.3.1 Introduction to TTP .............................................................................................. 6

1.2.3.2 von Willebrand Factor .......................................................................................... 6

1.2.3.3 ADAMTS13 ......................................................................................................... 7

1.2.4 Atypical Hemolytic Uremic Syndrome – a Complement-Mediated TMA ............ 8

1.2.4.1 A Brief Introduction of Hemolytic Uremic Syndrome – Typical and Atypical ... 8

1.2.4.2 The Complement System of Innate Immunity ...................................................... 9

1.2.4.3 Endothelial Cell Protection against Complement-Mediated Injury .................... 12

1.2.4.5 Complement-mediated „atypical‟ HUS............................................................... 13

1.2.4.6 CD46/MCP-associated atypical hemolytic uremic syndrome ............................ 14

1.2.5 From Mutation in CD46 to TMA – An Incomplete Story .................................... 15

1.2.5.1 Functional Studies of CD46/MCP Mutations ..................................................... 15

1.2.5.2 Incomplete Penetrance Suggests a Multiple „Hit‟ Concept ................................ 16

1.2.5.3 An Additional Complement Gene Aberrancy Increases Disease Penetrance ..... 16

1.2.5.4 A Link Between Complement and Coagulation ................................................. 17

1.3 Blood Outgrowth Endothelial Cells (BOEC) .................................................... 20

1.3.1 Introduction to BOEC ............................................................................................. 20

1.3.2 BOEC in the Study of Disease ................................................................................. 20

1.3.1 BOEC as a Model to Study TMA ........................................................................... 22

1.4 Knowledge Gap, Hypothesis and Thesis Aims .................................................. 23

Page 6: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

VI

CHAPTER 2 MODELING CD46/MCP-ASSOCIATED ATYPICAL

HEMOLYTIC UREMIC SYNDROME USING BLOOD OUTGROWTH

ENDOTHELIAL CELLS .......................................................................................... 24

2.1 Abstract 25

2.2 Introduction .......................................................................................................... 26

2.3 METHODS ........................................................................................................... 28

2.3.1 BOEC Isolation ........................................................................................................ 28

2.3.2 BOEC Characterization By Flow Cytometry ........................................................ 28

2.3.3 BOEC Characterization By Immunofluorescence ................................................ 29

2.3.4 Surface Expression Of Membrane-Bound Regulators On BOEC ....................... 30

2.3.5 Quantitative Gene Expression Of Membrane-Bound Regulators On BOEC .... 31

2.3.6 Complement Challenge Of BOEC .......................................................................... 32

2.3.6.1 50% normal human serum in alternative pathway buffer ................................... 32

2.3.6.2 Membrane-anchored complement regulator blockade ........................................ 32

2.3.6.3 Complement deposition detected by immunofluorescence ................................ 32

2.3.6.4 Complement deposition detected by FACS ........................................................ 33

2.3.7 Cell death assays ....................................................................................................... 34

2.3.7.1 Trypan blue exclusion ......................................................................................... 34

2.3.7.2 Cell cytotoxicity/LDH assay ............................................................................... 34

2.3.7.3 Apoptosis ............................................................................................................ 34

2.3.8 Platelet Adhesion ...................................................................................................... 35

2.3.8.1 Platelet isolation .................................................................................................. 35

2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ......................................... 35

2.4 RESULTS 37

2.4.1 BOEC Have An Endothelial Cell Phenotype And Retain Their Phenotype Over

Various Passages ............................................................................................................... 37

2.4.2 BOEC Express Key Membrane-Anchored Complement Regulators ................. 39

2.4.3 Cell Surface C3b Deposition Mirrors Incremental Functional Blockade of

CD46/MCP, CD55/DAF and CD59 ................................................................................. 41

2.4.4 Complement-mediated BOEC cytotoxicity increases with incremental

membrane-regulator blockade ......................................................................................... 43

2.4.5 Increasing complement challenge to BOEC results in an increase in platelet

adhesion.............................................................................................................................. 45

2.4.6 Discussion .................................................................................................................. 48

2.4.7 Supplemental Data ................................................................................................... 51

Page 7: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

VII

CHAPTER 3. VON WILLEBRAND FACTOR MULTIMERS CONTRIBUTE

TO COMPLEMENT ALTERNATIVE PATHWAY CONTROL ........................ 53

3.1 Abstract 54

3.2 Introduction .......................................................................................................... 56

3.3 Methods 58

3.3.1 Establishing and characterizing blood outgrowth endothelial cells (BOEC) ..... 58

3.3.2 Glomerular Endothelial Cell Culture ..................................................................... 58

3.3.3 Determining BOEC protein expression via Western blot .................................... 59

3.3.4 Quantifying BOEC and GEC Gene Expression via qRT-PCR ............................ 59

3.3.5 Characterization of membrane-bound regulators of BOEC ................................ 60

3.3.5.1 Immunofluorescence ........................................................................................... 60

3.3.5.2 Flow cytometry ................................................................................................... 61

3.3.6 Complement Challenge of BOEC ........................................................................... 62

3.3.6.1 Normal human serum.......................................................................................... 62

3.3.6.2 Membrane-anchored complement regulator blockade ........................................ 62

3.3.7 Assessment of Complement Challenge of BOEC .................................................. 63

3.3.7.1 Complement deposition detected by immunofluorescence ................................ 63

3.3.7.2 Complement deposition detected by FACS ........................................................ 63

3.3.8 Assessing the impact of complement challenge ..................................................... 64

3.3.8.1 LDH cell cytotoxicity assay ................................................................................ 64

3.3.8.1 Platelet adhesion Assay ...................................................................................... 64

3.3.9 Ethics ......................................................................................................................... 65

3.3.10 Statistical analysis .................................................................................................. 65

3.4 Results 66

3.4.1 BOEC Possess Endothelial Cell Characteristics ................................................... 66

3.4.2 Type 3 VWD BOEC Express Similar Amounts of Membrane-Anchored

Complement Regulators as Control BOEC .................................................................... 67

3.4.3 Complement activation products associate with VWF ......................................... 69

3.4.4 Platelet adhesion in response to complement challenge is initially dependent on

VWF release....................................................................................................................... 71

3.4.5 VWD BOEC Show Increased C3b Deposition after Complement Challenge .... 73

3.4.6 VWD BOEC Are More Vulnerable To Complement-Mediated Cytotoxicity .... 74

3.5 Discussion 76

3.6 Supplemental Data -VWF expression in glomerular endothelial cells ........... 79

Page 8: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

VIII

CHAPTER 4. UNIFYING DISCUSSION, FUTURE DIRECTIONS AND

CONCLUSIONS ........................................................................................................ 81

4.1 Unifying Discussion .............................................................................................. 82

4.2 Future Directions ................................................................................................. 86

4.2.1 Introduction .............................................................................................................. 86

4.2.2 Do Endothelial Cells Mount a Cytoprotective Response when Challenged by

Complement? ..................................................................................................................... 87

4.2.2.1 Rationale and Hypothesis ................................................................................... 87

4.2.2.2 Methods .............................................................................................................. 88

4.2.2.3 Preliminary Results ............................................................................................. 88

4.2.2.4 Discussion ........................................................................................................... 90

4.2.3 How Does the Fluid Phase Regulator CFH Contribute to Endothelial Cell

Protection? ......................................................................................................................... 91

4.2.3.1 Rationale and Hypothesis ................................................................................... 91

4.2.3.2 Methods .............................................................................................................. 91

4.2.3.3 Preliminary Results ............................................................................................. 92

4.2.3.3 Discussion ........................................................................................................... 94

4.2.4 Will BOEC Isolated From a Patient Further our Understanding of TMA? ...... 95

4.2.4.1 Rationale, Hypothesis and Aims ......................................................................... 95

4.2.4.3 Proposed Methods and Anticipated Results ....................................................... 96

4.2.4.4 Anticipated Results and Implications ................................................................. 96

4.3 Conclusions ........................................................................................................... 98

References 100

Page 9: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

IX

List of Tables

Table 1-1. Classification of Thrombotic Microangiopathies……………..……5

Table 1-2. Relevant Complement Regulators…………………………………14

Page 10: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

X

List of Figures

Figure 1-1. Overview of complement activation ...................................................... 11

Figure 2-1. BOEC have an endothelial cell phenotype and retain their phenotype

over various passages ................................................................................................. 39

Figure 2-2. BOEC express key membrane-anchored complement regulators ..... 41

Figure 2-3. Cell surface C3b deposition mirrors incremental functional blockade

of CD46/MCP, CD55/DAF and CD59 ...................................................................... 42

Figure 2-4. Complement-mediated BOEC cytotoxicity increases with incremental

membrane-regulator blockade ................................................................................. 44

Figure 2-5. Increasing complement challenge to BOEC results in a stepwise

increase in platelet adhesion ..................................................................................... 46

Figure 2-S1. Functional blocking antibody binding and saturation ...................... 51

Figure 2-S2. Expression of surface-bound regulators on glomerular endothelial

cells compared to BOEC ........................................................................................... 52

Figure 3-1. BOEC possess endothelial cell characteristics ...................................... 67

Figure 3-2. Type 3 VWD BOEC express similar amounts of membrane-anchored

complement regulators as control BOEC ................................................................ 68

Figure 3-3. Complement challenge results in VWF release and the association of

complement activation products and VWF ............................................................. 70

Figure 3-4. Platelet adhesion in response to complement challenge is initially

dependent on VWF release ....................................................................................... 72

Figure 3-5. VWD BOEC show increased C3b deposition after complement

challenge...................................................................................................................... 73

Page 11: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

XI

Figure 3-6. VWD BOEC are more vulnerable to complement-mediated

cytotoxicity .................................................................................................................. 75

Figure 3-S1. Glomerular endothelial cells express less VWF ................................. 79

Figure 4-1. VWF and ADAMTS13 as a Complement Regulatory System. ........... 85

Figure 4-2. Response of complement regulators to increasing complement

challenge...................................................................................................................... 90

Figure 4-3. Role of CFH-mediated surface protection against endothelial cell

death ............................................................................................................................ 93

Figure 4-4. Multiple genetic hits in complement and coagulation genes needed to

cause aHUS ................................................................................................................. 96

Page 12: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

XII

List of Abbreviations

ADAMTS13 A Disintergrin and Metalloprotease with Thrombospondin-1 Motifs

Type 13

ACD Acid Citrate Dextrose

AHUS Atypical Hemolytic Uremic Syndrome

AP Alternative Pathway

BOEC Blood Outgrowth Endothelial Cell

CC3 Cleaved Caspase 3

CFH Complement Factor H

CFI Complement Factor I

DAF Decay Accelerating Factor/CD55

EC Endothelial Cell

FBS Fetal Bovine Serum

HEPES Hyroxyethyl Piperazineethanesulfonic

MCL Mononuclear Cell Layer

MCP Membrane Cofactor Protein

MFI Median Fluorescence Intensity

NHS Normal Human Serum

PBS Phosphate Buffered Saline

TMA Thrombotic Microangiopathy

TTP Thrombotic Thrombocytopenic Purpura

VWF Von Willebrand Factor

Page 13: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

1

CHAPTER 1 AN INTRODUCTION TO THROMBOTIC

MICROANGIOPATHY

Page 14: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

2

A hidden connection is stronger than an obvious one.

Heraclitus of Ephesus (535–475 BC)

Page 15: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

3

1.1 Introduction

This introductory chapter will discuss the clinic-pathological entity thrombotic

microangiopathy (TMA). TMA is clinically manifest as anemia, thrombocytopenia

and acute kidney injury and pathologically, as swollen, damaged microvascular

endothelial cells coupled with platelet-rich microthrombi. There is an increasing

spectrum of diseases leading to TMA, and to this day, still a lack of clarity in

distinguishing where these diseases overlap and where they differ, in terms of

pathomechanisms. At the core of this thesis is an attempt to better understand a

particular form of TMA, atypical hemolytic uremic syndrome (aHUS), a disease

where a dysregulated alternative pathway (AP) of complement is thought to injure the

microvascular endothelium, thus creating a procoagulant nidus for the aggregation of

platelets. The AP of complement is a constitutively active cascade of intravascular

proteins that can ultimately lead to the formation of a pore-forming protein complex

in the plasma membrane of a cell and lysis of that cell. It can be rapidly amplified if

the cell surface is not recognized as being part of the host, which makes it an efficient

means of killing bacteria, or labeling them for phagocytosis and immune processing.

As such, it can be viewed as a sentinel of immune surveillance. Such a system needs

tight regulation and there are a number of regulators that can arrest the complement

cascade or allow for selective, i.e. site- and time- directed activation. Some of these

are soluble in the circulation, and capable of binding and inactivating complement

proteins that are either in the fluid phase or already attached to the host endothelium,

while others are membrane-anchored. Mutations in both the fluid-phase and

membrane-bound regulators have been linked to the pathogenesis of aHUS in about

two thirds of cases.(Noris and Remuzzi, 2009) Of note, complement regulatory

mutations have in general a low penetrance of about 50%. (Noris and Remuzzi, 2009)

Mutations in one of the endothelial cell-anchored regulators, CD46/Membrane

Cofactor Protein (MCP) have been found in about 10% of aHUS cases but these have

an even lower penetrance than some of the other mutations associated with aHUS,

such as the fluid phase regulator complement Factor H (CFH) for instance. There

remain significant gaps in our knowledge in explaining this variation, and in

understanding the path from a dysregulated complement system to a TMA phenotype.

Page 16: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

4

1.2 Thrombotic microangiopathy

1.2.1 Introduction to Thrombotic Microangiopathy

Thrombotic microangiopathy (TMA) is a pathological development arising from the

formation of intravascular thrombi, or clots, in the microvasculature, leading to a

microangiopathic hemolytic anemia, occlusion of the vessels and damage of the

tissues downstream of the occluding thrombi. (Kwaan, 2011) On a pathological level

there is capillary and arteriolar thrombosis accompanied by endothelial cell (EC)

swelling and damage.(George and Nester, 2014) Symmers proposed the term

thrombotic microangiopathy in 1952 to indicate “the location and the most striking

feature of the characteristic histological lesions without mentioning inconsistent and

controversial features of this clinico-pathological entity”. (Symmers, 1952) TMA is a

multisystem disease and can, in principle, affect any organ system including the heart,

lungs, brain, liver, pancreas, skin, bones and kidneys, with effects ranging from

sudden death to progressive organ damage and loss. The latter is a hallmark of kidney

TMA, especially in children, which affects the glomeruli, the blood-filtering vascular

units of the kidney. TMA can be an acute and life-threatening disease with serious

long-term sequelae and in some forms, a risk of recurrence. (Cataland and Wu, 2014;

Clark, 2012; Deford et al., 2013; Radhi and Carpenter, 2012)

1.2.2 The Spectrum of Thrombotic Microangiopathy

TMA is defined by the occurrence of occluding thrombi in the microvasculature here

is an increasing spectrum of diseases associated with a TMA and to date at least nine

separate TMA „syndromes‟ have been defined. (George and Nester, 2014; Riedl et al.,

2014a; Riedl et al., 2014b) Broadly speaking the TMAs can be categorized as being

either hereditary or acquired. (George and Nester, 2014) The principal causes and

associations of TMA are outlined in Table 1-1. Specific causes of TMA include: (i)

thrombotic thrombocytopenic purpura (TTP), (ii) hemolytic uremic syndrome (HUS),

(iii) drugs, such as cyclosporine or quinine, (Kojouri et al., 2001; Miller et al., 1997)

(iv) pregnancy-related TMA, (D'Angelo et al., 2009) (v) the inherited metabolic

disorders of cobalamin such as methyl malonic academia and homocystinuria (Van

Hove et al., 2002) and (vi), transplant associated TMA, particularly in association

with antibody-mediated rejection (Noone et al., 2012; Satoskar et al., 2010) and post

stem cell transplantation.(Jodele et al., 2014; Jodele et al., 2013) Although there is an

Page 17: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

5

emerging spectrum of diseases leading to a TMA, TTP and HUS are the two

quintessential diseases associated with TMA. (George and Nester, 2014)

Table 1-1: Classification of Thrombotic Microangiopathies Thrombotic thrombocytopenic purpura (TTP)

TMA associated with ADAMTS13 deficiency

Hereditary

Autoimmune

Infection induced TMA

(Enterohemorrhagic) E.coli- (EHEC)-associated HUS

Shigella dysenteriae HUS

Streptococcus pneumonia HUS

Influenza A/H1N1HUS

HUS due to other pathogens: EBV, CMV, Mycoplasma pneumoniae,

Bordetella pertussis, Parvovirus B19, HIV

Complement-Mediated ‘Atypical’ hemolytic uremic syndrome (aHUS)

Hereditary (mutations in CFH, CFI, MCP, C3 or CFB)

Autoimmune (autoantibodies to CFH)

TMA associated with pregnancy

TMA associated with transplantation

TMA developed de-novo after solid organ transplantation

TMA associated with bone-marrow transplantation or stem-cell

transplantation

TMA associated with metabolic disease

Cobalamin C deficiency

TMA associated with other glomerulopathies/vasculitides

Systemic lupus erythematosus/Antiphospholipid syndrome

C3 glomerulopathy (C3G)

Others: IgA nephropathy, focal segmental glomerulosclerosis (FSGS),

vasculitis

Drug induced TMA

Calcineurin inhibitors

Others: quinine, ticlopidine, chemotherapy

Other forms of TMA

DGKE mutation

Adapted from (George and Nester, 2014; Lemaire et al., 2013; Riedl et al., 2014a; Riedl et al., 2014b))

Page 18: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

6

1.2.3 Thrombotic Thrombocytopenic Purpura, VWF and ADAMTS13

1.2.3.1 Introduction to TTP

Eli Moschcowitz first described TTP in a 16-year-old girl who died within two weeks

of presenting with hemolytic anemia, thrombocytopenia, fever and neurological

symptoms. At post-mortem examination hyaline thrombosis of the capillaries was

found. (Moschcowitz, 1924) It was almost 60 years later when unusually large von

Willebrand Factor (VWF) multimers, a hemostatic protein, were identified in TTP

patients‟ plasma. (Moake et al., 1982) Sixteen years later the reason for the presence

of these unusually large VWF multimers in the plasma of TTP patients was

discovered to be a defect in the VWF cleaving protease ADAMTS13 (A Disintegrin

And Metalloproteinase with a ThromboSpondin type 1 motif, member 13). This was

either an acquired phenomenon via an inhibiting antibody or due to a mutation of

ADAMTS13. (Furlan et al., 1998; Tsai and Lian, 1998) TTP occurs either as the

hereditary Upshaw-Schulman syndrome (Rennard and Abe, 1979) where there are

either homozygous or compound heterozygous mutations in ADAMTS13, multimers,

or autoantibodies directed against it (Moschcowitz disease). (Kinoshita et al., 2001;

Levy et al., 2001; Sasahara et al., 2001; Tsai, 2013; Zheng et al., 2001)

Apart from the severe congenital form, Upshaw-Schulman syndrome, TTP is more

common in adults. TTP is manifest clinically with a non-immune microangiopathic

hemolytic anemia, thrombocytopenia, altered neurological status, kidney failure and

fever. This makes up the classically described pentad of TTP. (Knobl, 2014) Activity

levels of ADAMTS13 less than 10% may corroborate the diagnosis. (George and

Nester, 2014)

1.2.3.2 von Willebrand Factor

VWF is a glycoprotein contained as ultra large multimers in Weibel-Palade bodies

(WPB) of the ECs that are released upon cell activation. It is synthesized either in

ECs or megakaryocytes where it is stored in the WPB. VWF is also stored in the

granule of platelets. (Valentijn and Eikenboom, 2013) VWF is initially synthesized as

a monomer that dimerizes via its C-terminus and finally forms multimers through N-

terminal interactions. Multimers may either be released basally into the circulation or

trafficked to the WPB where the multimerization process continues, forming ultra

Page 19: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

7

large VWF (ULVWF) greater than 10,000 kDa in size. (De Ceunynck et al., 2013)

ULVWF can be released from a single WPB or from a „secretory pod‟ of coalesced

WPB. (De Ceunynck et al., 2013) Under the influence of blood flow, globular VWF

elongates into strings of between 100 -500 μm in length, (De Ceunynck et al., 2011)

which trap and bind platelets thus initiating clot formation. (De Ceunynck et al., 2013;

Nightingale and Cutler, 2013; Turner et al., 2009; Turner et al., 2012) VWF strings

anchored to ECs are much more efficient at adhering platelets after shear stress

exposes the A1 domain. (De Ceunynck et al., 2013; Schneider et al., 2007) VWF also

serves as a carrier protein for Factor VIII, protecting it from degradation and

clearance, thus further facilitating hemostasis. (Nightingale and Cutler, 2013)

1.2.3.3 ADAMTS13

It was initially recognized that VWF in the circulation was cleaved by a protease,

(Furlan et al., 1996; Tsai, 1996) and that this protease was a zinc protease, termed

ADAMTS13. (Levy et al., 2001; Zheng et al., 2001) ADAMTS13 prevents VWF

multimers from becoming excessively large by cleaving the VWF (Dong, 2005) at a

peptide bond in its A2 domain (Dong et al., 2003) and VWF multimers attached to the

EC undergo structural alterations under shear flow that enable ADAMTS13 to cleave

them. (Lancellotti and De Cristofaro, 2011; Lopez and Dong, 2004) When

ADAMTS13 is congenitally deficient or neutralized by an autoantibody, then

ULVWF and platelet rich thrombi may occlude the microcirculation. Platelets avidly

bind and are firmly fixed to ULVWF multimers that have been unfolded in the

circulation, thus forming a nidus for clot formation. (Levy et al., 2001; Tsai, 2010,

2013; Tsai and Lian, 1998; Tsai et al., 2006)

Page 20: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

8

1.2.4 Atypical Hemolytic Uremic Syndrome – a Complement-Mediated

TMA

1.2.4.1 A Brief Introduction of Hemolytic Uremic Syndrome – Typical and Atypical

Hemolytic uremic syndrome (HUS) is a life-threatening disease, typically of

childhood, where the child develops a TMA with anemia, thrombocytopenia, and

subsequent kidney failure after infection by an enterohemorrhagic, Shiga toxin-

producing E coli (STEC) strain. (Gasser et al., 1955; Karmali et al., 1983; Tarr et al.,

2005) About 15% of children infected with STEC will go on to develop HUS 7-10

days after ingestion of the bacteria and this is usually associated with bloody diarrhea.

The triad of a non-immune, microangiopathic hemolytic anemia, thrombocytopenia

and acute kidney injury characterizes HUS. Historically this was classified as

diarrhea-positive HUS and occasionally is associated with epidemics or outbreaks.

(Tarr et al., 2005) Although STEC HUS can present with a severe illness recovery is

usually spontaneous, there are no relapses and overall, the prognosis is good.

(Scheiring et al., 2008)

Following the recognition of HUS as a new disease entity came the realization that

there was a subtype of HUS, that was „atypical‟, in that it was either recurrent (Brain,

1969; Kaplan, 1977) or familial and had a more severe presentation and worse

outcome. (Hagge et al., 1967; Kaplan et al., 1975) These atypical forms were

recognized to occur after a nonspecific prodrome and were diarrhea negative. (Kaplan,

1977) Early in the history of atypical HUS, disturbance and activation of the

complement system was noted. (Barre et al., 1977; Carreras et al., 1981; Drukker et

al., 1975) It had also been noted that plasma therapy could replace some, as yet

unknown and presumed deficient plasma factor. (Remuzzi et al., 1978; Remuzzi et al.,

1979) Furthermore, plasma exchange was effective in cases where infusion seemed

insufficient. (Camba et al., 1985; Remuzzi and Ruggenenti, 1992) Low levels of the

complement regulator Factor H (CFH) had been described in aHUS patients, (Pichette

et al., 1994; Thompson and Winterborn, 1981) but the major breakthrough came when

Warwicker et al mapped the inherited form of HUS to the region on chromosome 1q

that encoded complement Factor H (CFH) and confirmed genetic deficiency in this

complement regulator as being the cause. (Warwicker et al., 1999; Warwicker et al.,

Page 21: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

9

1998b) Warwicker and Goodship were the first to propose a “complement-based

theory of microangiopathy”. (Warwicker et al., 1998a)

1.2.4.2 The Complement System of Innate Immunity

Complement proteins form part of the innate immune system where they

„complement‟ and augment antibody-mediated bacterial killing, linking innate and

adaptive immunity, as well as protect the host from various infectious challenges and

aid in the clearance of immune complexes and cellular debris. (Walport, 2001a)

Origins of the complement system can be traced back to 500 million years

ago.(Nonaka, 2014) There are three principal complement activation pathways,

namely the classical (CP), mannose-binding lectin (MBL) and AP.

The classical pathway is activated when C1q, part of the C1 protein complex that

includes the serine proteases C1s and C1r, binds antigen-antibody complexes of either

IgG or IgM, resulting in the cleavage of C4 into C4a and C4b. C4b can then bind C2.

The serine protease C1s cleaves the C4b-bound C2 into C2a and C2b. The remaining

C4bC2a acts as a C3 convertase, splitting it into the anaphylatoxin C3a, and C3b.

C4bC2aC3b acts as a C5 convertase to cleave C5 into the anaphylatoxin C5a, and

C5b, thus initiating the terminal complement cascade (TCC). C5b deposited on cell

surfaces associate with the proteins C6, C7, C8 and C9 to form the heteropolymeric,

membrane attack complex, C5b-9 (MAC). (Degn and Thiel, 2013; Matsushita et al.,

2013; Muller-Eberhard, 1986)

In the mannose-binding lectin pathway, polysaccharides and sugars on the surface of

pathogens are bound by either mannose binding lectin (MBL) or ficolins. The MBL

also have associated serine proteases (MASP 1 and 2) that are phylogenetically

analogous to the C1 complex, and capable of cleaving and activating both C4 and C2.

(Matsushita et al., 2013; Wallis, 2007)

The alternative pathway is constitutively active, amplifiable and targets any

unprotected surface. (Gotze and Muller-Eberhard, 1976) This is in contrast to the CP

and MBL that are activated by binding certain pathogen association molecular

patterns. Spontaneous activation of C3 occurs in the fluid phase via a „tick-over

mechanism‟ related to instability of the thioester bond within the C3 alpha chain. C3

in which the thioester bond has been exposed and hydrolyzed will naturally associate

with H20. Complement Factor B can bind C3(H20) to form C3(H20)B. Factor D

Page 22: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

10

subsequently cleaves C3(H20)B, thereby forming the AP C3 convertase C3(H20)Bb

and initiating the AP. (Degn and Thiel, 2013; Ricklin et al., 2010) Properdin stabilizes

the AP convertase thus enhancing amplification of this pathway. (Fearon and Austen,

1975; Muller-Eberhard, 1988; Pillemer et al., 1954) C3b(H20)Bb cleaves more C3

creating an amplification loop and the AP C5 convertase, C3bBbC3b, is formed

allowing progression to the TCC and construction of C5b-9/MAC. (Pangburn et al.,

2008) It has recently been observed that the AP can act as an amplifier of the other

two pathways, increasing the amount of C5 cleaved and C5b-9 formed by up to 80%.

(Harboe and Mollnes, 2008; Harboe et al., 2004)

No matter how the complement cascade is activated, all three pathways converge at

the level of hydrolysis (i.e. activation) of C3, the most abundant complement protein

in plasma, with the formation and deposition of C3b onto any target cell surface. C3

activation can occur both in the fluid phase and on surfaces. The attachment of C3b to

the cell surface is followed by the formation of a C3 convertase (C3bBbP), capable of

cleaving more C3. Within seconds to minutes a cell surface can be coated in C3b.

(Pangburn et al., 2008) When C3b (like C4b) is bound to an antigen or cell it can label

it for processing by antigen-presenting cells or for phagocytosis (opsonization).

Alternatively the terminal cascade of complement can be initiated and propagated by

the C3 and C5 convertases, leading to the formation of the membrane attack complex,

C5b-9. (Pangburn et al., 2008; Walport, 2001a) The deposition of C3b on cells is

indiscriminate, and recognition and protection of the host is achieved principally by a

system of regulators, discussed below. (Pangburn et al., 2008)

Page 23: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

11

Figure 1-1. Overview of complement activation

Complement may be activated via the classical, mannose-binding lectin or the APs. All

pathways converge at the level of C3. The classical pathway is activated by the C1 protein

complex of C1q and the serine proteases C1s and C1r. Mannose-associated serine protease

(MASP) 1 & 2 are involved in the initiation of the MBL pathway. The AP is continuously

active and amplifiable. All three converge at the level of C3 and have a common terminal

pathway to formation of the membrane attack complex, C5b-9. (Walport, 2001b)

Recently, a fourth pathway of activation was recognized which, in contrast to the

other pathways, bypasses C3 activation. Thrombin, a serine protease that catalyzes the

conversion of fibrinogen to insoluble fibrin strands to aid coagulation, has been

recognized as this fourth, C3 independent, pathway of complement activation.

(Huber-Lang et al., 2006) Furthermore, thrombin can generate a unique C5 split

(C5bT) product that when acted on by the C5 convertase results in a MAC complex

with increased lytic activity. (Krisinger et al., 2012) Thrombin can also cleave C3 to

form the anaphylatoxin C3a. (Amara et al., 2008)

Page 24: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

12

1.2.4.3 Endothelial Cell Protection against Complement-Mediated Injury

Over 60 trillion (1012

) endothelial cells make up the largest interconnected organ in

the human body, the lining of the vasculature, covering as much as 4,000 square

meters. (Aird, 2007a) Host protection of the vascular endothelium is extremely

important, especially against the AP of complement, an innate system of immune

surveillance that is constitutively active, amplifiable and ends with a membrane lysing

protein complex. In fact, all human cells have complement regulators on their surface.

(Zipfel and Skerka, 2009) Regulation of complement occurs with a combination of

fluid phase regulators, including complement Factor H (CFH) and membrane-

anchored regulators including CD35/CR1, CD46/Membrane Cofactor Protein (MCP),

CD55/Decay Accelerating Factor (DAF) and CD59. These are outlined in table 1-2.

(Campbell et al., 2002; Skidgel and Erdos, 2007; Thurman and Renner, 2011;

Tschopp and French, 1994) The overarching functional principle of these regulators is

the interference with the progression of the complement cascade at the level of C3b or

the C3 convertase. CFH is a cofactor for the serine protease complement Factor I that

cleaves and inactivates C3b. It also serves to accelerate the decay of the C3

convertase and as a host recognition molecule. (Ferreira et al., 2010) On the EC

surface CD55/DAF also accelerates the decay of the C3 convertase, while CD59

prevents formation of C5b-9 by binding C8 and C9. (Thurman and Renner, 2011)

Within the kidney, glomerular endothelial cells (GEC) express CD46/MCP,

CD55/DAF and CD59, (Endoh et al., 1993; Ichida et al., 1994; Nakanishi et al., 1994)

but do not express the EC surface regulator CR1. (Thurman and Renner, 2011).

CD46/MCP is a transmembrane protein identified first in 1985 as a C3b binding

protein expressed on human mononuclear cells. (Cole et al., 1985) As stated above,

CD46/MCP binds C3b (and C4b) and has cofactor activity for CFI, allowing for

cleavage and inactivation of the complement activation product C3b deposited on the

host cell surface. (Seya and Atkinson, 1989; Seya et al., 1986) Originally described

and characterized on blood cells, (Seya et al., 1988) and later on EC, (McNearney et

al., 1989) CD46/MCP is coded for by a gene lying within the region of regulators of

complement activation (RCA) gene cluster on chromosome 1q32. (Liszewski et al.,

1991) Within the kidney, CD46/MCP expression is ubiquitous, on glomerular

structural cells, podocytes and tubular epithelial cells. (Endoh et al., 1993; Ichida et

al., 1994; Nakanishi et al., 1994) Its role as a complement regulator was extensively

Page 25: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

13

studied (Barilla-LaBarca et al., 2002; Brodbeck et al., 2000; Liszewski and Atkinson,

1996) prior to its discovery to be related to aHUS some 15 years later.(Richards et al.,

2003) Structurally, CD46/MCP consists of an extracellular domain with four

complement control protein modules, and an O-glycosylated region rich in serines,

threonines and prolines termed the “STP” region, a cytoplasmic anchor and tail.

Alternative splicing of the CD46 gene affects both the “STP” region and cytoplasmic

tail resulting in principally four isoforms of CD46/MCP. (Post et al., 1991) The

cytoplasmic tail may be responsible for directing the intracellular protein to the cell

surface. (Maisner et al., 1996)

1.2.4.5 Complement-mediated ‘atypical’ HUS

Atypical HUS (aHUS) can be either sporadic or familial, and has been linked to

defective regulation of the innate immune and coagulation systems. (Delvaeye et al.,

2009; Kavanagh et al., 2006; Warwicker et al., 1998b) Atypical HUS (Noris et al.,

2010) is an early-onset disease where many patients have been identified as having

hereditary (e.g. mutations) and/or acquired (e.g. autoantibodies) predispositions

related to the regulation of the complement AP (AP), which involves both plasma-

borne factors (e.g. factor H; CFH) and proteins on the surface of vascular ECs (e.g.

Membrane Cofactor Protein CD46/MCP). (Malina et al., 2012) Different from STEC

HUS, atypical HUS has a high risk of progression to end stage kidney disease (>50%)

and death (up to 25%), or the need for lifelong dialysis or extremely expensive

treatments because these patients are poor candidates for transplant due to disease

recurrence in the graft. (Noris and Remuzzi, 2009) Children with aHUS have a five

times greater mortality as compared to adults.(Fremeaux-Bacchi et al., 2013) Atypical

HUS has been linked to mutations in several genes, detectable in about 60% of cases.

(Caprioli et al., 2006; Noris et al., 2010) Most of these genes are in the regulator of

complement activation (RCA) region of Chr 1 (1q32). (Rodriguez de Cordoba et al.,

1999) Loss of AP regulation results in the generation of excessive amounts of

anaphylatoxins (C3a, C5a), opsonins (C3b) and membrane attack complexes

(MAC/C5b-9) both in fluid phase and on vascular ECs in glomeruli, (Waters and

Licht, 2011) where damage releases signals promoting inflammation and platelet

Page 26: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

14

activation leading to thrombus/clot formation. The resultant TMA in the kidney

microvasculature causes to in acute kidney injury (aHUS). (Zipfel et al., 2006)

Table 1-2: Relevant Complement Regulators

Regulator Function

Fluid Phase Regulators

CFH Accelerates decay of C3 convertase

Cofactor for the serine protease CFI that

cleaves C3b into inactive form iC3b

C4b binding protein Functions as CFH but for the CP

Vitronectin/ S protein Binds to C5b-7 to prevent C5b-9

formation

Clusterin Binds to C5b-7 to prevent C5b-9

formation

Carboxypeptidase N Inactivates the anaphylatoxins C3a &

C5a

Pro-carboxypeptidase R

(proCPR)/thrombin-activatable

fibrinolysis inhibitor (TAFI)

Inactivates the anaphylatoxins C3a &

C5a

Membrane-Anchored Regulators

CD46/Membrane Cofactor Protein Cofactor for the serine protease CFI that

cleaves C3b into inactive form iC3b

CD55/Decay Accelerating Factor Accelerates decay of C3 convertase

CD59 Binds C5b-8 on cell membrane

preventing C5b-9 formation

CD35/CR1 Cofactor for the serine protease CFI that

cleaves C3b into inactive form iC3b &

Accelerates decay of C3 convertase

1.2.4.6 CD46/MCP-associated atypical hemolytic uremic syndrome

The principal EC bound complement regulator first recognized as linked to aHUS is

membrane cofactor protein (MCP/CD46). (Pirson et al., 1987; Richards et al., 2003)

CD46 gene mutations have been found in up to 10% of aHUS patients.(Fremeaux-

Bacchi et al., 2013; Fremeaux-Bacchi et al., 2006) The mutations in MCP related

aHUS are mainly heterozygous, compound heterozygous or homozygous and

translate into a reduced cell surface expression of the CD46/MCP protein in more

Page 27: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

15

than 75% of cases. (Fremeaux-Bacchi et al., 2013; Goodship, 2006) This reduced cell

surface expression is due to retention of the immature precursor protein within the cell.

(Goodship, 2006) The remainder of MCP mutations result in a protein expressed at

normal levels but lacking either cofactor activity or C3b binding capacity. (Esparza-

Gordillo et al., 2005; Noris et al., 2003; Richards et al., 2003) Studies in transfected

Chinese hamster ovary cells have found that this reduced expression translates into

reduced regulation of the AP, (Liszewski et al., 2007) ultimately then thought to lead

to enhanced C5b-9/MAC generation.

Although those with CD46/MCP related aHUS occur at a younger age and have a

higher rate of recurrence than those with other forms of aHUS including CFH related

aHUS, they are three to four times more likely to retain normal renal function and

have 75% long-term renal survival. (Fremeaux-Bacchi et al., 2013; Noris et al., 2003;

Noris et al., 2010; Sellier-Leclerc et al., 2007) CD46/MCP related aHUS is somewhat

unique, in that its rate of recurrence post-transplant is minimal, likely owing to the

fact that the kidney allograft will be expressing non-mutant CD46/MCP. (Caprioli et

al., 2006; Fremeaux-Bacchi et al., 2007; Richards et al., 2007)

1.2.5 From Mutation in CD46 to TMA – An Incomplete Story

1.2.5.1 Functional Studies of CD46/MCP Mutations

Functional studies of CD46/MCP mutations associated with aHUS have been

conducted using Chinese Hamster Ovary (CHO) cell transfection, and CHO cells

transfected with MCP mutations were demonstrated to having an increased cell

surface deposition of C3b. However, CHO cells express no other complement

regulators and, therefore, studies performed in CHO cells do not take into account the

contribution of any of the other complement regulators that would be present, and

presumably functional, in vivo, on human EC. Thus, although an excellent way for

assessing the functional impact of individual MCP mutations (Brodbeck et al., 2000;

Liszewski and Atkinson, 1996) (Liszewski et al., 2008; Liszewski et al., 2007) this

system does not allow for interpreting the role of MCP mutations in context of the

physiological setting of multilayered complement regulators e.g. on EC with respect

Page 28: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

16

to the manifestation of a TMA phenotype. For the most part it is assumed that this

increase in C3 deposition implies progression to the TCC with more C5b-9

production, EC activation, injury or death and the emergence of a prothrombotic EC

surface upon which platelets aggregate. (Noris and Remuzzi, 2009). However, this

pathogenetic sequence of TMA has never been fully proven and hinges significantly

on assumptions.

1.2.5.2 Incomplete Penetrance Suggests a Multiple ‘Hit’ Concept

Mutations in CD46/MCP associated with aHUS – like other aHUS causing mutations

– are not automatically disease causing but are associated with variable penetrance.

(Dragon-Durey and Fremeaux-Bacchi, 2005) In fact a sibling-pair both harboring a

homozygous deletion in CD46/MCP and lacking CD46/MCP has been reported where

one has recurrent aHUS and the other absolutely no disease.(Couzi et al., 2008) This

incomplete penetrance is not unique to CD46/MCP, and in other forms of

complement-mediated aHUS, a patients‟ family members may also carry the same

mutation and remain unaffected.(Kavanagh et al., 2005; Martinez-Barricarte et al.,

2008; Noris et al., 2010) This variable, or incomplete penetrance, suggests a threshold

for the development of aHUS. It has been suggested that mutations in the complement

system may serve as predisposing factors for the development of aHUS, where some

mutations may be associated with a greater risk. Then, with a certain „trigger‟ event –

either environmental or provided by another genetic defect – the patient may manifest

the disease. Infection seems to be a common trigger event, where children often

present after a nonspecific prodrome suggestive of a viral illness, or have a disease

recurrence seemingly precipitated by an intercurrent illness. (Johnson and Waters,

2012) Other triggers preceding aHUS manifestation include immunization, pregnancy,

surgical operations etc. (ref – if at hand)

1.2.5.3 An Additional Complement Gene Aberrancy Increases Disease Penetrance

Having a combination of mutations in complement genes has been described in aHUS.

(Bienaime et al., 2010; Geerdink et al., 2012; Maga et al., 2010; Noris et al., 2010;

Sellier-Leclerc et al., 2007) In an American cohort, 12% of the patients had mutations

in more than one gene, (Maga et al., 2010) and a relatively small pediatric cohort of

45 children with aHUS found 4 with combined mutations. (Geerdink et al., 2012)

Page 29: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

17

Furthermore, certain haplotypes and single nucleotide polymorphisms in complement

genes, genetic variations in DNA or single nucleotides that are very close and

therefore inherited alongside each other, have been identified in aHUS patients and

may act as disease susceptibility factors. (Fremeaux-Bacchi et al., 2005) These

variations can be additive, for instance in one pedigree only individuals possessing

three - a mutation and SNP in CD46/MCP as well as an insertion in the gene for CFI

– developed aHUS. (Esparza-Gordillo et al., 2006) SNPs alone in CD46 are likely

insufficient to cause the disease and a mutation is also required.(Ermini et al., 2012)

Recent evidence has shown that the combination of complement gene „risk haplotypes‟

can increase the disease penetrance. (Bresin et al., 2013) The presence of both a CFH

and CD46 risk haplotype increases disease penetrance to 73% versus 36% among

carriers with zero or one risk haplotype. It also seems that as well as increasing

disease penetrance, the natural course of the disease may also be altered by having a

combination of haplotypes as 50% of patients with combined CD46 mutation

developed ESRD within 3 years from onset as compared to just 19% of patients with

an isolated CD46 mutation in this study.(Bresin et al., 2013)

1.2.5.4 A Link Between Complement and Coagulation

The coagulation and complement systems have been partners in protecting their hosts

for millions of years, although a greater understanding of the extent of the interaction

between the two systems continues to evolve. (Delvaeye and Conway, 2009;

Markiewski et al., 2007; Oikonomopoulou et al., 2012) The convergence of both

systems has recently come to the foreground, especially with the finding of an

additional C3-independent pathway of complement activation by thrombin, a

proteolytic serine protease integral to coagulation. (Huber-Lang et al., 2006) This was

followed by the finding that thrombomodulin (THBD; CD141), a cofactor for

thrombin, also functioned as a cofactor for CFI-mediated C3 cleavage, therefore

acting as a complement regulator and that certain heterozygous missense mutations

could be linked to patients with aHUS. (Delvaeye et al., 2009) Furthermore,

pathogenic variants in coagulation genes, notably PLG, that encodes plasminogen,

were recently found in an aHUS cohort. (Bu et al., 2014)

Page 30: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

18

Thrombotic microangiopathy (TMA) is a pathological process where this complex

interplay between the complement system and coagulation has always been prominent

and somewhat controversial, especially in terms of distinguishing TTP and aHUS.

Both clinically and pathologically it can be difficult to distinguish the two. (Fujimura,

2003; Remuzzi, 1987, 2003; Tsai, 2003) As it became evident that the underlying

cause was clearly different, related to defective complement regulation in aHUS, and

failure of ADAMTS13 to cleave VWF multimers in TTP, the two seemed clearly

distinct.(Tsai, 2003) However more recently it has emerged that complement may

play a role in TTP, blurring the lines of separation again. (Noris et al., 2012; Reti et

al., 2012; Ruiz-Torres et al., 2005; Wu et al., 2013; Zipfel et al., 2011) Sera from TTP

patients can activate the AP leading to EC cytotoxicity in vitro. (Ruiz-Torres et al.,

2005) Reti et al demonstrated that the anaphylatoxin C3a and soluble C5b-9 levels

were higher in TTP patients during acute disease. (Reti et al., 2012) Following on

from that, Wu et al analyzed stored blood samples from a cohort of 38 TTP patients,

10 of whom died, for complement activation. Thirty three of 38 (87%) had evidence

of complement activation at presentation and in those that died, there was evidence of

both AP and terminal complement cascade activation with levels of Bb, C3a, C5a and

C5b-9 all significantly higher in this group. (Wu et al., 2013)

Recently, the link between complement and coagulation in aHUS has been expanded

to involve VWF and its cleaving protease, ADAMTS13. In a recent study involving

29 aHUS patients, Feng et al found additional ADAMTS13 polymorphisms

associated with decreased ADAMTS13 activity. (Feng et al., 2013a) 80% of the

cohort also carried at least 1 non-synonymous change in ADAMTS13, and in 38% of

patients, multiple ADAMTS13 variations were found. These ADAMTS13 variants

were likely significant as measured ADAMTS13 activity was less than 60% in half of

the patients studied.(Feng et al., 2013a) Alongside this clinical observation, there have

been a number of experimental studies linking CFH, ADAMTS13 and VWF. Turner

and Moake demonstrated the assembly and activation of AP components, especially

C3 and C5, on ultra large VWF strings secreted from and anchored to ECs.(Turner

and Moake, 2013) CFH has been shown to be a reductase for large soluble VWF

(LsVWF) multimers that are secreted from ECs. (Nolasco et al., 2013) Large soluble

VWF multimers released from complement activated ECs assume a conformation that

makes them inaccessible to ADAMTS13 cleavage, but they can still bind platelets and

Page 31: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

19

induce platelet aggregation under conditions of high-fluid shear stress. (Nolasco et al.,

2013) The C-terminus of CFH has been shown to enhance ADAMTS13 VWF

cleavage in vitro. (Feng et al., 2013c) Finally, CFH cofactor activity may be enhanced

by an interaction between VWF and CFH. (Rayes et al., 2014) Putting this all together,

it has recently been postulated that the association between VWF and the AP of

complement may be complement amplifying, which would be very pertinent to TMA

and aHUS. (Turner et al., 2014)

Page 32: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

20

1.3 Blood Outgrowth Endothelial Cells (BOEC)

1.3.1 Introduction to BOEC

It has recently become feasible to isolate endothelial progenitor cells (EPCs) from

circulating peripheral blood mononuclear cells (PBMC), and culture them to obtain

“outgrowth” populations known as BOEC (Lin et al., 2000) or endothelial colony-

forming cells (ECFCs). (Reinisch et al., 2009) BOEC or endothelial colony-forming

cells (ECFCs), are considered by some authors to be true EC precursors, having a

distinct transcript signature compared to early endothelial progenitor cells (EPCs) and

a high proliferative potential. Populations of these cells can be expanded in multiple

passages for 3-4 months without them undergoing any transformation and with

retention of their EC traits in terms of genotype and phenotype.(Fuchs et al., 2010;

Fuchs et al., 2006; Timmermans et al., 2009) Their precursor has not yet been

identified and it is possible that they originate from blood vessels in vivo, as they are

phenotypically similar to mature ECs, yet have a much greater proliferative

capacity.(Tura et al., 2013)

1.3.2 BOEC in the Study of Disease

BOEC have proven useful in studies of vascular disease; e.g. transcriptome analysis

detected upregulation of disease-relevant genes in BOEC from patients with

proliferative diabetic retinopathy, whose cells also showed diminished migratory

capacity and integration into retinal endothelium. (Tan et al., 2010) BOEC expression

array analysis detected several genes with roles in endothelial biology that may

influence susceptibility to shear stress and cardiovascular disease. (Ahmann et al.,

2011; Ensley et al., 2012; Mazzolai et al., 2011) BOEC are also well suited to in vitro

modeling of cell interactions and pathological conditions such as high shear stress and

ischemia-reperfusion. These cells can be obtained with high efficiency from small

volumes of peripheral blood using cell separation methods and direct culture, and they

have close affinities to their donor‟s vascular ECs. (Martin-Ramirez et al., 2012;

Page 33: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

21

Reinisch et al., 2009) For example BOEC have been used to elucidate the functional

symbiosis between glomerular endothelium and epithelium mediated by podocyte-

secreted VEGF. (Hirschberg et al., 2008) They have also proven useful in studies

relating to thrombotic disease. In a shear stress model BOEC were observed to alter

their cytoskeletal structure and antithrombogenic potential, (Ensley et al., 2012) and

showed changes in expression of receptors for tissue plasminogen activator, urokinase

and adhesion molecules. (Ahmann et al., 2011; Mazzolai et al., 2011) BOEC have

also been used to study the effects mutations in EC-expressed genes (e.g. von

Willebrand Factor [VWF] deficiency(Othman et al., 2010)), and there is evidence that

BOEC and other EPCs may represent populations that play important roles in

recovery from thrombotic disease (e.g. stroke(Yang et al., 2012)). Several groups

have begun to explore the therapeutic potential of EPCs for vascular cell

transplantation(Kaneko et al., 2012) (with or without genetic modification) and

antithrombotic endothelialization. (Kaneko et al., 2012; Yoon et al., 2005) BOEC can

also be used to study the functional effects of a known EC mutation as has been done

by Othman et al, where they characterized the functional significance of a specific

von Willebrand Factor (VWF) mutation. (Othman et al., 2010) BOEC have been used

to study the effects of mutations in EC-expressed genes (e.g. von Willebrand Factor

[VWF] deficiency. (Othman et al., 2010; Wang et al., 2013b)

BOEC represent a potentially valuable source of material for gene expression analysis

and the search for disease-associated gene variants using a transcriptomic approach.

(Fernandez et al., 2005; Medina et al., 2010b; Tan et al., 2010) The application of

global microarray profiling of BOEC from patients with a defined genetic phenotype

can provide insight into the mechanisms underlying that disease at the level of the EC.

(Medina et al., 2010a) Different subjects might exhibit different combinations of

polymorphisms that affect EC gene expression and biologic systems. (Enenstein et al.,

2010) Chang Milbauer et al studied BOEC lysates from sickle cell disease patients at

risk for stroke. (Chang Milbauer et al., 2008) By defining a priori, defined biologic

systems that could be implicated in this risk at the endothelial level (e.g. angiogenesis,

hypoxia response, coagulation, shear stress response and inflammation) (Chang

Milbauer et al., 2008) and then assembling specific gene sets to survey that system,

went on to identify enhanced activity of each biologic system. Of note early

expansion stages of BOEC may reveal a transiently acquired phenotype, especially if

Page 34: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

22

taken from patients with active inflammation or disease;(Kahlenberg et al., 2011) for

e.g. in burn patients more BOEC colonies were isolated as compared to normal

controls and these BOEC secreted more VEGF than normal counterparts. (Rignault-

Clerc et al., 2012) However, Chang Milbauer et al have shown that after a 10-fold

expansion in the number of cells (at least one passage), some upregulated genes return

back to normal expression levels. (Chang Milbauer et al., 2008) Thus the „acquired

phenotype‟ may be washed out and expanded BOEC reflect culture conditions and the

inherent genetics of the subject‟s ECs. These cells can also be cryopreserved. A study

has shown that after two years cryopreservation at -80 degrees Celsius, almost 80% of

the cells remained viable.(Wagner and Myrup, 2005)

1.3.1 BOEC as a Model to Study TMA

To date most experimental studies on the pathogenesis of TMA and aHUS have used

EC lines such as human umbilical vein ECs, glomerular EC lines (Frimat et al., 2013;

Louise and Obrig, 1994; Ray et al., 2006) or CHO cells. (Liszewski et al., 2007) The

use of non-endothelial cell-based systems however has limitations; (i) it cannot

entirely recapitulate the physiological endothelial milieu and (ii) there are problems

with overexpression/underexpression of the relevant gene being studied, and (iii) the

effect on the other regulators cannot be easily studied. BOEC are an endothelial

progenitor cell that offer a number of advantages over cell lines such as HUVECs and

offer the unique opportunity to directly study patient specific ECs where they will

reflect the particular genotype of that particular individual.

Page 35: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

23

1.4 Knowledge Gap, Hypothesis and Thesis Aims

The principal gap in knowledge revolves around explaining the steps from loss of

complement regulation on the endothelium in association with a mutation in

CD46/MCP, to the manifestation of a TMA phenotype of EC death and platelet

microthrombi. What is known comes from studies in CD46/MCP-transfected CHO

cells, a non-human cell, unrelated to the endothelium, and devoid of other

complement regulators. Published, disease-causing mutations in CD46/MCP are less

efficient at inactivating C3b deposited on complement-challenged CHO cells.

Thereafter, it is assumed that the terminal complement cascade gets activated with

more C5b-9 formation, EC injury leading to a procoagulant surface with platelet

aggregation. The fact that the disease is associated with variable penetrance suggests

additional „hits‟ may be required.

Hence, the specific hypotheses tested are the following:

1. In any individual carrying a functionally relevant CD46/MCP mutation, an

additional complement regulatory defect acts as a second „hit‟ and increases

the likelihood of TMA manifestation.

2. Complement-induced EC activation and VWF release acts as an additional „hit‟

via amplification of the complement cascade.

The primary aim of this thesis is to examine how loss of complement regulation on

the EC surface leads to a thrombotic microangiopathic phenotype in an ex vivo model

of disease that incorporates human blood outgrowth endothelial cells (BOEC), an

endothelial progenitor cell that can ultimately be grown from patients, and an

endothelialized microfluidic system.

The secondary aim is to evaluate the contribution of von Willebrand Factor (VWF) to

the pathogenesis of atypical hemolytic uremic syndrome (aHUS).

Page 36: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

24

CHAPTER 2 MODELING CD46/MCP-ASSOCIATED

ATYPICAL HEMOLYTIC UREMIC SYNDROME USING

BLOOD OUTGROWTH ENDOTHELIAL CELLS

Page 37: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

25

2.1 Abstract

Atypical hemolytic uremic syndrome (aHUS) is due to defective regulation of the

alternative complement pathway (AP), which is constitutively active and requires a

multi-layered defense system. It remains unclear how this defense system functions as

a whole to protect injured endothelial cells (EC) or why loss/diminished function of

one of these regulators results in disease. Mutations in CD46/membrane cofactor

protein (MCP), a membrane-anchored CAP regulator leads to aHUS but with variable

penetrance. We aimed to use blood outgrowth endothelial cells (BOEC), endothelial

cell precursors that can be isolated from the peripheral blood, to study aHUS

pathogenesis ex vivo. BOECs were cultured from subjects by a standard protocol. EC

phenotype and presence of complement regulators was confirmed by

immunofluorescence (IF), western blot (WB), qPCR, and flow cytometry (FACS).

Cells were challenged with complement, with or without functional blockade of

complement regulators. Cell death and apoptosis were quantified. Calcein-labelled

platelets were perfused across confluent BOEC in a microfluidic system and platelet

adhesion was quantified. BOEC were characterized by FACS as being positive for the

endothelial cell markers CD31/PECAM-1 and CD144/VE-cadherin, while being

negative for the hemangioblast marker CD45 and immature endothelial progenitor

cell marker, CD14. Surface expression of complement regulators was confirmed. A

surrogate phenotype of aHUS modelled ex vivo (endothelial cell death and platelet

adhesion) is not achieved with CD46/MCP blockade alone. Complement deposition

(C3) is increased by blocking complement regulators simultaneously (four-fold

increase when CD46/MCP, CD55/DAF and CD59 are blocked; p <0.001) and results

in cell death, apoptosis and an EC prothrombotic phenotype with platelet adhesion.

We have established an ex vivo method to model aHUS pathogenesis that can now be

expanded to use patient-derived BOEC. Our data supports the hypothesis that an

additional factor is required in order for a patient with a CD46/MCP mutation to

manifest disease.

Page 38: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

26

2.2 Introduction

Thrombotic microangiopathy (TMA) is a disease affecting the microvasculature,

especially within the kidney, characterized by endothelial cell injury and occlusion of

capillaries and arterioles with platelet microthrombi. Atypical hemolytic uremic

syndrome (aHUS) is one form of TMA typically with a young age of onset that can be

caused by either inherited, or an acquired, dysregulation of the constitutively active

complement alternative pathway (AP). Genetic causes / mutations giving rise to

aHUS either result in loss of appropriate AP regulation by fluid phase (complement

Factors H and I, CFH and CFI respectively), or membrane-anchored regulators

CD46/membrane cofactor protein (MCP), or amplification of the AP by mutant C3 or

complement Factor B (CFB).(Noris and Remuzzi, 2009)

The principal endothelial cell -anchored complement regulator first recognized as

linked to aHUS is CD46/MCP.(Pirson et al., 1987; Richards et al., 2003) CD46/MCP

binds C3b and has cofactor activity for the serine protease complement Factor I (CFI),

allowing for cleavage and inactivation of the complement activation product C3b

deposited on the host cell surface.(Seya and Atkinson, 1989; Seya et al., 1986)

Mutations in CD46/MCP are found in up to 10% of aHUS patients.(Fremeaux-Bacchi

et al., 2006) Mutations are mainly heterozygous, but homozygous, and null mutations

have also been described and involve reduced expression, impaired C3b binding or

retention of a CD46/MCP precursor within the cell. CD46/MCP related aHUS is

somewhat unique, in that unlike the other forms of aHUS, its rate of recurrence post-

transplant is minimal owing to the fact that the kidney allograft will be expressing

non-mutant CD46/MCP.(Caprioli et al., 2006; Fremeaux-Bacchi et al., 2007;

Richards et al., 2007)

How an individual mutation leads to a TMA phenotype and aHUS is not always clear.

Studies of transfected Chinese hamster ovary (CHO) cells have found that reduced

expression or loss of CD46/MCP translates into reduced regulation of the AP and

more C3b deposition on the cell surface.(Liszewski et al., 2007) This might be

expected to translate in vivo, on the human microvascular endothelium, into more

membrane attack complex/C5b-9 (MAC/C5b-9) formation, endothelial cell activation,

Page 39: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

27

injury or death and a prothrombotic endothelial cell phenotype with platelet adhesion

and a TMA. However, as clinically affected probands often have unaffected siblings

or relatives carrying the same mutation, current opinion proposes a

„multiple/cumulative-hit‟ sequence of events leading up to disease precipitation.

Disease may become manifest only with the presence of an additional mutation or risk

haplotype, for instance as combination of CFH and CD46/MCP mutations, which has

been shown to (almost) double disease penetrance over carrying individual mutations

and to be associated with a more severe disease and worse outcome.(Bresin et al.,

2013)

Blood outgrowth endothelial cells (BOEC), are an endothelial progenitor that can be

isolated with high efficiency from small volumes of peripheral blood using cell

separation methods and direct culture, and they have close affinities to their donor‟s

vascular endothelial cells.(Martin-Ramirez et al., 2012; Schiff et al., 2004) Although

phenotypically similar to mature endothelial cells, they have a much greater

proliferative capacity and retain their endothelial cell traits in terms of genotype and

phenotype across many passages.(Fuchs et al., 2006; Timmermans et al., 2009; Tura

et al., 2013) BOEC can be used to study the functional effects of known endothelial

cell-expressed genes, with studies in von Willebrand Factor (VWF) deficient patients

and von Willebrand Disease being most successful to date.(Othman et al., 2010;

Wang et al., 2013a)

In this study, using BOEC, we model the impact of a functional defect in CD46/MCP

and show that loss of function of CD46/MCP alone may be insufficient to result in a

TMA phenotype of increased complement deposition, endothelial cell death and

platelet adhesion.

Page 40: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

28

2.3 METHODS

2.3.1 BOEC Isolation

48 ml of whole blood was obtained from healthy adult donors in six cell preparation

tube (CPT) vacutainers (Becton Dickinson, Franklin Lakes, USA). Tubes were spun

at 1600 x g for 30 minutes to obtain the serum and mononuclear cell layer (MCL).

The serum and MCL were added to 8 ml of 10% fetal bovine serum (FBS: Sigma-

Aldrich, St. Louis, USA) in phosphate buffered saline (PBS: Wisent). Cells were

pelleted at 520 x g and resuspended in 10% FBS in PBS twice. Using Trypan Blue

(Sigma-Aldrich, St. Louis, USA) and an hematocytometer (Hausser Scientific,

Horsham, USA) the number of cells were quantified. Cells were pelleted at 520 x g

and resuspended in cEGM-2 media with supplements (Lonza, Walkersville, USA: Cat

no. 362753), 10% FBS and 1% Antibiotic-Antimycotic (Gibco, Invitrogen, Life

Technologies, Carlsbad, USA; containing 10,000 units/mL of penicillin, 10,000

µg/mL of streptomycin, and 25 µg/mL of Fungizone® Antimycotic). 3-5 x 107 cells

were aliquoted to each well in a six-well tissue culture plate (Becton Dickinson,

Franklin Lakes, USA) coated with 0.05 mg/ml of rat tail collagen type I in 0.02 M

acetic acid (Becton Dickinson, Franklin Lakes, USA). Cells were kept at 37°C and in

an environment with 5% CO2. For the first 7 days after seeding the cells, 3.5 mL of

media is aspirated carefully, 2 ml of pre-warmed media is gently added, and then

carefully aspirated before replacing with 4 mL of media. Thereafter media is replaced

once every two days for approximately two weeks to a month until other blood cells

die off and a monolayer of BOEC are identifiable by their endothelial cobblestone

morphology and ready for passaging (Figure 2-1A).

2.3.2 BOEC Characterization By Flow Cytometry

Alexa Fluor 488 conjugated mouse anti-human CD31 (PECAM-1) [5μL/test], PE

conjugated mouse anti-human CD144 (VE-Cadherin) [12μL/test], Alexa Fluor 647

conjugated mouse anti-human CD14 [5μL/test] and APC-H7 conjugated mouse anti-

human CD45 [3μL/test] were used for surface staining of the BOEC, with Alexa

Fluor 488 mouse IgG2 κ, PE mouse IgG1 κ, Alexa Fluor 647 mouse IgG2b κ and

APC H7 mouse IgG1 κ, as isotype controls. Cells were stained with Fixable Viability

Dye eFluor 450 (eBioscience, San Diego, USA), fixed with 2% PFA/PBS and

Page 41: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

29

blocked with 200 μg/mL final volume of purified mouse serum IgG (Sigma-Aldrich,

St. Louis, USA) prior to specific or isotype antibody incubations. A minimum of

60000 live cells were collected using a Beckman Coulter Gallios flow cytometer

(Beckman Coulter, Brea, USA), equipped with 4 excitation lasers (405nm, 488nm,

561nm, 633nm). Data was analyzed with Kaluza software (Beckman Coulter).

HUVEC passage 6 were used as a positive control for the endothelial cell markers

CD31 (PECAM-1) and CD144 (VE-Cadherin) and blood as a positive control for

CD45 and CD14. Fluorescence minus one (FMO) controls using BOEC were stained

for each of the four specific antibodies and were used as the gating controls during

data analysis. Antibodies were purchased from BD Pharmingen (San Diego, USA).

2.3.3 BOEC Characterization By Immunofluorescence

BOEC at about 80-90% confluence were washed once with HBSS (Gibco, Life

Technologies, Carlsbad, USA), trypsinized (with 0.05% trypsin/0.53mM EDTA,

WISENT, St Bruno, Canada) then resuspended in cEGM-2 (BOEC) media to a

concentration of 0.5 × 106 cells/ml. 500 μL of the BOEC suspension is added to 2.5

mL of cEGM-2 (BOEC) media already pre-warmed in each well of a 6-well tissue

culture dish containing previously collagen-coated 22x22-mm cover slips (VWR

International, Radnor, USA). BOEC adhere to the cover slips after overnight

incubation (37°C, 5% CO2). Samples were washed with ice-cold PBS then fixed with

4% (w/v) paraformaldehyde (Electron Microscopy Sciences, Fort Washington, USA)

in PBS. Samples were blocked with 2% (w/v) donkey serum (Jackson

ImmunoResearch, West Grove, USA) either alone or with 0.2% (v/v) Triton X-100

(for permeabilization) for 60 minutes. Samples were stained overnight with the

relevant primary antibodies made up in the relevant blocking solution. Samples were

washed in PBS and incubated with respective donkey-anti secondary antibodies with

Alexa Fluor® 488 conjugate (Invitrogen, Life Technologies, Carlsbad, USA). Cell

nuclei were stained by either DAPI (1 μg/ml) or 0.12 μg/ml Hoechst stain (Thermo

Fisher Scientific, Waltham, USA) for ten minutes. Samples were washed in PBS and

cover slips were mounted with Dako Fluorescence Mounting Media (Dako, Glostrup,

Denmark) for analysis with either a Nikon Eclipse Ti microscope (Leika

Microsystems, Wetzlar, Germany) or spinning disk confocal microscopy. The latter is

equipped with an Olympus IX81 inverted fluorescence microscope using a 60./1.35

Page 42: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

30

oil immersion objective equipped with a Hamamatsu C9100-13 back-thinned EM-

CCD camera and Yokogawa CSU X1 spinning disk confocal scan head (with upgrade

from Spectral Aurora Borealis, Richmond Hill, Canada). The unit is equipped with 4

separate diode-pumped solid state laser lines (Spectral Applied Research, Richmond

Hill, Canada, 405 nm, 491 nm, 561 nm, and 642 nm) with emission filters: 447 nm ±

60, 525 nm ± 50, 593 nm ± 40, 620 nm ± 60, 676 ± 29 and 700 nm ± 75, and 1.5X

magnification lens (Spectral Applied Research). Confocal images were taken with an

Improvision Piezo Focus Drive. Z-stacks were taken at 0.25 μm. Images taken using

the spinning disk confocal microscope were deconvolved by iterative restoration

using Volocity Software (PerkinElmer, Waltham, USA) with confidence limit set to

95% and iteration limit set to 20.

The following antibodies were used for IF: sheep anti-human VWF (1:1000 dilution,

Abd Serotec, Oxford, UK, AHP062), goat anti-human P-selectin (CD62P, 1:100

dilution; Santa Cruz Biotechnology, Dallas, USA; sc-6943), rabbit polyclonal

antibody to MCP (1:50 dilution; Santa Cruz Biotechnology, Dallas, USA; sc-9098,),

rat polyclonal antibody to CD59 (1:1000 dilution; Abd Serotec, Oxford, UK;

MCA715G), and goat polyclonal antibody to CD55 (1:50 dilution; R&D Systems,

Minneapolis, USA; AF2009). Nuclei of cells were stained with 0.12 μg/ml Hoechst

stain (Thermo Fisher Scientific, Waltham, USA) for ten minutes. All secondary

antibodies utilized were conjugated to either Alexa Fluor 488 or Alexa Fluor 555

(Life Technologies, Carlsbad, USA) dyes.

2.3.4 Surface Expression Of Membrane-Bound Regulators On BOEC

Cells were seeded overnight in a 6 well plate (Falcon, Becton Dickinson, Franklin

Lakes, USA) to confluence, washed 1x with HBSS and then trypsinized (0.05%

Trypsin/0.53mM EDTA, WISENT). The primary antibodies (anti-CD46, Santa Cruz

Technology, Dallas, USA, sc-9098; anti-CD55, R&D, Minneapolis, USA, AF2009;

anti-CD59, AbD Serotec, Oxford, UK, MCA715G) and secondary antibodies (Alexa

fluor 488, Invitrogen, Life Technologies, Carlsbad, USA) were incubated at 4° for 20

minutes. Primary antibodies were used at a dilution of 1:100, secondary antibodies at

a 1:200 dilution. Twenty thousand events were recorded using Attune Acoustic

Focusing Cytometer (Life Technologies, Carlsbad, USA) and analyzed with FlowJo

Page 43: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

31

software (FlowJo LLC, Ashland, USA). Results are given as median fluorescence

intensity (MFI). Surface expression was compared with that of glomerular endothelial

cells in passage 6.

2.3.5 Quantitative Gene Expression Of Membrane-Bound Regulators On

BOEC

Cells (BOEC and HUVEC) were seeded overnight in a 6 well plate (Falcon) to

confluence, washed 1x with HBSS before RNA was isolated using TRI Reagent

(Sigma-Aldrich, St. Louis, USA, T9424) according to manufacturer‟s instructions.

RNA concentration and integrity was verified by spectrophotometer (NanoDrop 1000,

Thermo Fisher Scientific, Waltham, USA), and reverse transcribed using

ReadyScript™ cDNA Synthesis Mix (Sigma-Aldrich, St. Louis, USA, RDRT).

Samples (200 ng cDNA in diethyl pyrocarbonate (DEPC, Sigma-Aldrich) treated

water) were amplified by real time polymerase chain reaction (PCR) using StepOne™

System from Life Technologies (Carlsbad, USA). Amplified products were detected

using KiCqStart™ SYBR® Green qPCR ReadyMix™, with ROX™ (Sigma-Aldrich,

Carlsbad, USA, KCQS02) and analyzed as follows: 2–(C

T – C

T GAPDH) – C

T control)

.

The following oligonucleotide primers (Sigma-Aldrich, Carlsbad, USA) were used:

GAPDH: forward, 5‟-ACAGTTGCCATGTAGACC-3‟; reverse, 5‟-

TTTTTGGTTGAGCACAGG-3‟.

VWF: forward, 5‟-TGTATCTAGAAACTGAGGCTG-3‟; reverse, 5‟-

CCTTCTTGGGTCATAAAGTC-3‟.

CD46/MCP: forward, 5‟-AGTGGTCAAATGTCGATTTC-3‟; reverse, 5‟-

ATCCCAAGTACTGTTACTGTC-3‟.

CD55/DAF: forward, 5‟-CAGAGGAAAATCTCTAACTTCC-3‟; reverse, 5‟-

AGTTGGTGAGACTTCTGTAG-3‟.

CD59: forward, 5‟-CATTACCAAAGCTGGGTTAC-3‟; reverse, 5‟-

TTTCTCTGATAAGGATGTCCC-3‟.

Page 44: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

32

2.3.6 Complement Challenge Of BOEC

2.3.6.1 50% normal human serum in alternative pathway buffer

Serum was collected from whole blood of adult donors into serum vacutainers

(Becton Dickinson, Franklin Lakes, USA), allowed to clot for 30 minutes and then

centrifuged at 3000 x g at 4°C. Serum was collected and stored at -20°C until needed

as a source for complement. Heat-inactivated serum (HIS), for use as a negative

control, was obtained by incubating the serum for 30 minutes at 56°C.

2.3.6.2 Membrane-anchored complement regulator blockade

In order to simulate a CD46/MCP mutation, a monoclonal mouse anti-human anti-

CD46 antibody (GB24, IgG1, kindly provided by John Atkinson, St. Louis, USA) was

used.(Turner et al., 1996) GB24 efficiently and completely blocks C3b binding and

cofactor activity of CD46/MCP in in vitro assays by binding to the complement

control proteins (CCPs) 3 and 4 of CD46/MCP. (Liszewski et al., 2000; Turner et al.,

1996) Additional blockade of endothelial cell surface complement regulation was

achieved with the use of a monoclonal anti-human CD55/DAF (BRIC216, IgG1) and

monoclonal anti-human CD59 (BRIC229, IgG2b) (International Blood Group

Reference Laboratory, NHS Blood and Transplant, Bristol, UK) functional blocking

antibodies. These antibodies are known to be non-complement activating. Antibodies

were used at a concentration of 5μg/mL (derived from antibody titration experiments

performed by FACS, Supplemental figure 2-S1) and diluted in serum free cEGM-2

(Lonza) media for 30 minutes in all experiments. To ensure antibodies were targeting

the correct receptors, BOECs were grown on 22x22 mm coverslips (VWR) coated

with 0.05 mg/ml of rat tail collagen type I (Becton Dickinson) in six well tissue

culture plates, fixed with 4% paraformaldehyde (Electron Microscopy Sciences) in

PBS and stained with the blocking antibodies and with one of the following

antibodies: goat anti CD55/DAF (R & D Systems), rat anti CD59 (AbD Serotec) or

rabbit anti CD46/MCP (Santa Cruz). (Supplemental figure S1)

2.3.6.3 Complement deposition detected by immunofluorescence

For IF demonstration of complement deposition (C3 and C5b-9), BOEC were grown

on collagen-coated coverslips and then incubated for 60-240 minutes (37°C, 5% CO2)

Page 45: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

33

with either cEGM-2 (BOEC) media, 50% normal human serum (NHS) in AP buffer

alone or after complement regulator blockade. After 60-240 minutes the supernatant

was aspirated and the cells were washed 3 times with ice cold PBS, blocked with 1%

(w/v) BSA for 1h and incubated with rabbit polyclonal antibody to C3c, (1:1000

dilution; Abcam, Cambridge, UK, ab15980) or a rabbit polyclonal antibody to C5b-9

(1:1000 dilution; Abcam, Cambridge, UK, ab55811) overnight.

2.3.6.4 Complement deposition detected by FACS

C3b deposition was demonstrated by FACS using a polyclonal rabbit anti-human

antibody to C3c (Abcam, ab15980) that detects C3c as well as the C3c part of native

C3 and C3b. Cells were seeded overnight in a 6 well plate (Falcon) to confluence,

washed 1x with HBSS before blocking antibodies (GB24, BRIC216, BRIC229) were

added for 20 minutes in serum-free media, followed by 50% NHS in AP buffer

(20mM HEPES pH7.4, 144mM NaCl, 7mM MgCl2, 10mM EDTA) for 1 hour. Cells

were trypsinized (0.05% Trypsin/0.53mM EDTA, WISENT) and washed in Flow

buffer (FB, 1% FBS/PBS) twice. The primary antibody in FB and the secondary

antibody (R-Phycoerythrin-conjugated AffiniPure F (ab‟)2 Fragment Donkey Anti-

Rabbit IgG (H + L), Jackson ImmunoResearch Laboratories, West Grove, USA, 709-

116-149, 1:200) together with Fixable Viability Dye eFluor780 (eBioscience, San

Diego, USA, 65-0865, 1:1000) in PBS were incubated at 4° for 20 minutes. At least

ten thousand events of BOEC population were recorded using Attune Acoustic

Focusing Cytometer (Life Technologies) and analyzed using FlowJo software.

Results are given as median fluorescence intensity (MFI). Cells were gated for live

cells (red laser 536nm, emission channel 2), single cells (FSC-A vs. FSC-H) and

finally through forward scatter and side scatter to determine the BOEC population.

C3c was recorded via the blue laser 488nm, emission channel 2. To correct for

spectral overlap during multicolor flow cytometry experiments, color compensation

was performed each time.

Page 46: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

34

2.3.7 Cell death assays

2.3.7.1 Trypan blue exclusion

BOECs (between passages 3-14) were grown in 0.05 mg/ml rat-tail collagen type I

coated 96 well tissue culture plates (Sarstedt, Nümbrech, Germany). Depending on

the treatment, cells were incubated for thirty minutes at 37°C with one of anti-

CD46/MCP, anti-CD55/DAF, anti-CD59, or all three regulator functional blocking

antibodies in serum free EGM-2 media with supplements (Lonza) as described in the

BOEC Isolation section. The controls were incubated with media only. Following

incubation, cells were exposed to either media or 50% normal human serum from

adult donors in AP buffer. This treatment lasted thirty minutes at 37°C. Then cells

were washed twice with phosphate buffered saline (PBS: Wisent, St Bruno, Canada).

1:1 mixture of Trypan Blue (Sigma-Aldrich, St. Louis, USA) in PBS was added to

cells for five minutes. 4% paraformaldehyde in PBS was utilized to fix cells for ten

minutes. Two fields of cells at 10x magnification from each of duplicate wells were

counted by using a Leitz DM IL microscope (Leica Microsystems). The values for

dead cells were calculated by finding the percentage of overall cells that were dead.

Standard deviation was calculated to create error bars and the student‟s t-test (two-

tailed) was utilized to determine significance (p<0.05).

2.3.7.2 Cell cytotoxicity/LDH assay

BOEC were seeded overnight in a 96 well ELISA plate (Sarstedt) to confluence,

washed 1x with HBSS (Gibco) before complement-blocking antibodies was added for

30 minutes in serum-free media (cEGM-2). After washing twice, 10% NHS in serum-

free media was added for 4 or 6 hours accordingly. Pierce LDH cytotoxicity assay kit

(Thermo Fisher Scientific, Waltham, USA) was used according to manufactures

instructions. OD was normalized to positive control and displayed in percent.

2.3.7.3 Apoptosis

Cells were seeded in 96-well ELISA plates (Sarstedt) and incubated with functional

blocking antibodies for 30 minutes followed by 50% NHS in serum-free media

(cEGM-2) for four hours. Cells were fixed and blocked as described earlier and

Page 47: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

35

stained for cleaved caspase-3 (Cell Signaling, Danvers, USA, 1:400 dilution) and

respective secondary antibody conjugated with Alexa fluor 555.

2.3.8 Platelet Adhesion

2.3.8.1 Platelet isolation

For platelet isolation, whole blood was collected from healthy adult donors and mixed

in a 6:1 ratio with acid citrate dextrose (ACD: 22.9 mM citric acid, 44.9 mM sodium

citrate dehydrate, 74 mM dextrose monohydrate). Platelet rich plasma (PRP) was

collected after centrifugation at 160 x g for ten minutes. PRP was spun at 950 x g for

seven minutes to pellet platelets. Platelets were washed twice with PBS/ACD solution

(20% ACD in PBS; pH 6.1). Platelets were incubated with 2.5 μM calcein (Life

Technologies, Carlsbad, USA) for thirty minutes at 37°C Finally, platelets were

pelleted at 950 x g and resuspended at a concentration of 15x107/ml in Tyrodes buffer

(136 mM NaCl, 2.7 mM KCl, 0.42 mM NaH2PO4, 19 mM NaHCO3, 5.5 mM of

glucose, 1 mM CaCl2, 1 mM MgCl2 and 10 mM of hyroxyethyl

piperazineethanesulfonic acid (HEPES: Invitrogen, Life Technologies, Carlsbad,

USA)). Platelet count was measured using an automatic hematocytometer (Beckman

Coulter, Brea, USA).

2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System

For BOEC-Platelet Interaction experiments, BOEC (passage 3-14) were seeded into

the BioFlux System (Fluxion Biosciences, South San Francisco, USA) channels the

day before the actual experiment. BOECs were grown in BioFlux 48 well tissue

culture plates coated with 0.05 mg/ml rat-tail collagen type I (Becton Dickinson). The

collagen was added to the output well and flown backward at a shear rate of

3dyne/cm2 for 1-2 minutes after which the plate was incubated overnight at 37°C.

BOEC were trypsinized and up concentrated to 8 x106 cells/mL. After adding 50 μl of

cEGM-2 to the input well as a balance, 50 μl of the cell solution were transferred to

the output well and then perfused backwards for 15 seconds at 1 dyne/cm2. After

sufficient cell adhesion was observed under the microscope, the plate was put into the

37°C incubator for 1 hour before adding 1 ml of cEGM-2 to each input well. Cells

were left in a 37°C / 5% CO2 incubator overnight. Cells were washed at 1 dyne/cm2

Page 48: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

36

with Hank‟s balanced salt solution (HBSS). Complement challenge was achieved by

flowing 50% NHS in alternative pathway (AP) buffer over the cells either, (i) alone,

(ii) after cells were pretreated with an anti-CD46/MCP function blocking antibody, or

(iii) after cells were pretreated with an antiCD46/MCP, anti-CD55/DAF and anti-

CD59 functional blocking antibodies. Blocking antibodies were flown over the cells

in serum free EGM-2 media (Lonza) while incubated at 37°C for thirty minutes

before washing with HBSS. 50% NHS was flowed through the BioFlux chambers at 2

dynes/cm2 for 1-2 hours. For platelet adhesion assays, 100 x 10

6 calcein (Life

Technologies) labeled platelets in Tyrodes buffer (as described in platelet preparation

section) were flowed through the chambers at 2 dyne/cm2 after BOEC exposure to

serum. After 5 to 10 min three pictures per channel were taken with a Leitz DM IL

microscope (Leica) at a magnification of 4x. Adhering platelets were counted using

ImageJ software.

2.3.9 Ethics Approval

Ethics approval was obtained from the Research Ethics Board at The Hospital for

Sick Children, Toronto, ON, Canada.

2.3.10 Statistical analysis

Figures were generated using GraphPad Prism 6 and displayed as mean and standard

deviation. Statistical analysis was performed using either two-way ANOVA with

post-hoc analysis or paired t-test. A p value < 0.05 was considered as statistically

significant. In figures p values are presented as follows: * ≤0.05, ** ≤0.01, ***≤0.001,

****≤0.0001, ns > 0.05.

Page 49: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

37

2.4 RESULTS

2.4.1 BOEC Have An Endothelial Cell Phenotype And Retain Their

Phenotype Over Various Passages

BOEC were isolated from the peripheral blood of four healthy adult donors. BOEC

appeared after a mean of 9 (range 7-20) days and displayed a typical cobblestone

appearance when visualized by light microscopy (Figure 2-1 A). An endothelial

phenotype was confirmed by demonstrating the presence of Weibel-Palade bodies

containing fluorescently labelled VWF and P-selectin (CD62P) imaged by confocal

spinning disk immunofluorescence microscopy (Figure 2-1 C, 2-1 D). BOEC can be

confirmed as late outgrowth endothelial colony forming cells as distinct from early

endothelial progenitor cells by the presence of specific endothelial cell markers and

the absence of certain surface markers.(Timmermans et al., 2009; Timmermans et al.,

2007) By flow cytometry cells were positive for the endothelial cell markers

CD31/PECAM-1 (platelet-endothelial cell adhesion molecule-1), and CD144 (VE-

Cadherin) [HUVECs as positive controls] and negative for the hemangioblast marker

CD45 and the immature EPC marker CD14 [blood as negative control]), consistent

with a BOEC phenotype (Figure 2-1 B). In order to confirm the stability of the cell

line across passages we showed that VWF expression by qPCR was the same at an

early (passage 3) and a late passage (passage 10) (Figure 2-1 E).

Page 50: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

38

Page 51: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

39

Figure 2-1. BOEC have an endothelial cell phenotype and retain their phenotype over

various passages

(A) BOEC are derived from mononuclear cells in peripheral blood that are cultured in cEGM-

2 medium. During the first week early endothelial progenitors (eEPCs) and fibroblasts are

seen. From week 2-3, blood outgrowth endothelial cells appear and form a typical

„cobblestone‟ monolayer as the eEPCs die off. (B) An endothelial phenotype was confirmed

by demonstrating the presence of Weibel-Palade bodies containing fluorescently labeled von

Willebrand factor (VWF, Alexa fluor 555; red) and P-selectin (CD62P, Alexa fluor 488;

green). We used an Olympus IX81 inverted fluorescence microscope using a 60/1.35 oil

immersion objective and a Hamamatsu C9100-13 back-thinned EM-CCD camera with

Yokogawa CSU X1 spinning disk confocal scan head. Confocal images were taken with an

Improvision Piezo Focus Drive. Z-stacks were taken at 0.25 μm. Images taken using the

spinning disk confocal microscope were deconvolved by iterative restoration using Volocity

Software with confidence limit set to 95% and iteration limit set to 20. (C) By flow cytometry

we further confirmed their endothelial phenotype, as BOEC are positive for PECAM/CD31

and VE-Cadherin/CD144 and negative for the hemangioblast marker, CD45 and immature

EPC marker, CD14. (D) By quantitative PCR we confirmed that BOECs are stable at an

early (passage 3) and late passage (passage 10) for one endothelial cell marker (VWF).

mRNA expression was normalized to GAPDH and displayed relative to VWF

expression of passage 3. N=2.

2.4.2 BOEC Express Key Membrane-Anchored Complement Regulators

We next wanted to confirm the presence of the key membrane-anchored complement

regulators on BOEC and compare this to HUVEC, a primary endothelial cell line used

in the study of aHUS pathomechanisms. The presence of these regulators

(CD46/MCP, CD55/DAF, CD59 and CD141/Thrombomodulin) was demonstrated by

using confocal, spinning disk microscopy of immunofluorescently labeled BOEC

grown to confluence on coverslips (Figure 2-2 A-C). The baseline gene expression

levels of CD46/MCP, CD55/DAF and CD59 were similar to that of HUVEC, when

assessed by qPCR (Figure 2-2 D). In both HUVECs and BOECs, there is a

significantly higher level of CD59 as compared to the other regulators CD46/MCP

Page 52: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

40

and CD55/DAF. This gene expression pattern was replicated by flow cytometry

assessment of cell surface expression of the proteins, where BOEC demonstrated a

similar amount of CD46/MCP and CD55/DAF (p=0.8) and a 14-fold higher relative

amount of CD59 on their surface (as compared to CD46; p=0.002) (Figure 2-2 E-F).

This was confirmed in three different control BOEC lines. The expression of

CD46/MCP, CD55/DAF and CD59 was also similar to that found on glomerular

endothelial cells. (Supplementary figure 2-S2)

Page 53: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

41

Figure 2-2. BOEC express key membrane-anchored complement regulators

(A-C) Complement regulators CD46/MCP (A), CD55/DAF (B) and CD59 (C) were

fluorescently labeled and imaged on BOEC by confocal spinning disk microscopy (see Figure

1 for details). (D) By qPCR we compared the relative expression of complement regulators as

compared to VWF (VWF expression=1). * p<0.05. Results were compared to expression

levels in HUVEC. No significant difference was observed (two-way ANOVA). N=6 for

HUVEC and N=5 for BOEC. (E-F) By flow cytometry we quantified a 14-fold higher

expression of CD59 (green) compared to CD46/MCP (blue) and a 9-fold higher expression

compared to CD55/DAF (orange) in three different control BOEC lines. No significant

difference was observed between surface expression of CD46/MCP and CD55/DAF. ** p<

0.01 compared to CD46 and CD55 (paired t-test). Median fluorescence intensity (MFI) was

calculated as fold increase over unstained control. N=3 In (E) one representative flow

cytometry reading of control BOECs is shown.

2.4.3 Cell Surface C3b Deposition Mirrors Incremental Functional

Blockade of CD46/MCP, CD55/DAF and CD59

The functional relevance of aHUS-associated CD46/MCP mutations has, to date, been

studied in a Chinese hamster ovary cell model system. When CHO cells that have

been transfected with the mutant protein are incubated with normal human serum, a

measurable increase in C3b deposition is demonstrated by flow cytometry, as

compared to wild type CD46/MCP expressing cells. (Goodship et al., 2004;

Liszewski et al., 2007; Richards et al., 2007) By contrast, we employed a model

system of BOEC, as these cells physiologically express membrane-bound

complement regulators, in contrast to CHO cells. 50% NHS (in AP buffer) was added

to confluent BOEC for a one hour period, either alone or after functional blockade of

the membrane regulators. Serum was removed; the cells were washed with HBSS,

then trypsinized and taken into suspension for FACS. Cells were labeled with a

polyclonal rabbit anti-human antibody to C3c and showed no significant increase in

C3 deposition over that seen by incubating the cells with 50% NHS alone when

CD46/MCP complement regulatory function was blocked (Figure 2-3 A-B, p>0.05).

Additional blockade of CD55/DAF and CD59 resulted in a 7-fold increase of C3c

deposition (p<0.05). On immunofluorescence an increase in C5b-9 deposition was

visualized when three surface-bound regulators were blocked and cells were perfused

Page 54: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

42

with 50% NHS for one hour in the BioFlux at a rate of 2 dynes/cm2, as compared to

single regulator blockade (Figure 2-3 D-F).

Figure 2-3. Cell surface C3b deposition mirrors incremental functional blockade of

CD46/MCP, CD55/DAF and CD59

(A-B) C3 deposition was evaluated by flow cytometry using an anti-C3c antibody (detects C3,

C3b, C3c but not C3a and C3d). (A) shows a representative figure, with the following

conditions: unstained (grey), 50% NHS (light green), CD46/MCP blocked prior 50% NHS

(dark green) and CD46/55 and 59 blocked prior adding 50% NHS (red) for one hour. In

Page 55: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

43

figure B the median fluorescence intensity (MFI) of 7-8 different experiments is shown

(*p<0.05, paired t-test).

(C-E) BOEC, cultured in a microfluidic chamber were perfused with 50% NHS for 1h.

BOEC in channel D were pretreated with an anti-CD46/MCP antibody and cells in channel E

with an anti-CD46/MCP, anti-CD55 and anti-CD59 antibody for 30 minutes. An increasing

C5b-9 deposition (polyclonal rabbit anti human C5b-9, 1:1000, secondary antibody: Alexa

fluor 555, 1:1000) was observed only when CD46/MCP, CD55/DAF and CD59 were blocked.

VWF was stained using a polyclonal sheep anti-human antibody (1:1000, secondary antibody:

Alexa fluor 488) and DNA using Hoechst dye (0.12μg/ml). Images were taken on oil-

immersion 60x magnification with a Nikon Eclipse Ti camera.

2.4.4 Complement-mediated BOEC cytotoxicity increases with

incremental membrane-regulator blockade

Current understanding of aHUS pathogenesis suggests that a dysregulated

complement AP can cause injury to the microvascular endothelium leading to EC

death and retraction. (Noris and Remuzzi, 2009) BOEC death was first assessed by a

Trypan blue exclusion assay. Confluent cells were subjected to an increasing

complement challenge with 30 minutes exposure to 50% NHS either alone, or after

functional blockade of the membrane regulators CD46/MCP, CD55/DAF and CD59.

Single regulator blockade failed to result in an increase in the percentage of dead cells

over that which occurs with 50% NHS alone. Intensifying the complement challenge

by the simultaneous blockade of three regulators, followed by exposure to 50% NHS,

resulted in a statistically significant increase in cell death (p=0.04) (Figure 2-4 A).

The same result was replicated when measuring LDH release after incubation with

10% NHS for 4 and 6 hours. Due to the high amount of LDH in serum this assay

could only be performed supplementing 10% serum to serum-free media (Figure 2-4

B). Furthermore, dead cells at 4 hours post complement challenge stained positive on

immunofluorescence for cleaved caspase-3 implicating apoptosis as the principal

mode of endothelial cell death after complement challenge at this timepoint (Figure 2-

4 C-E).

Page 56: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

44

Figure 2-4. Complement-mediated BOEC cytotoxicity increases with incremental

membrane-regulator blockade

(A) BOEC grown to confluence in a 96-well plate were exposed to 50% NHS for 30 minutes

after blocking complement regulators. Trypan-blue was added and cells counted on a Leitz

DM IL microscope (Leica) with a 10x magnification. Per well 2 random fields were counted,

two wells per permutation for a total of 4 assays. A significant increase of dead cells was seen

after blocking CD46/CD55/CD59 when compared to cells treated only with 50% NHS. N=4,

*p=0.04 (paired t-test). (B) This was confirmed by measuring release of LDH after incubating

confluent BOEC with 10% NHS in serum-free media for four and six hours, N=5 (blockade

of CD46/CD55/CD59 versus 50% NHS alone; p=0.03 and blockade of CD46/CD55/CD59

versus CD46 blockade; 0.02 respectively) (C-D) After incubation with 50% NHS for four

hours cells were fixed and stained for cleaved caspase 3 (1:400) and donkey anti rabbit Alexa

fluor A555 (red). DNA was stained using Hoechst dye (0.12μg/ml, blue). Cells were grown in

96-well plates for images taken at 20x magnification. (C) Representative image after

pretreatment with anti-CD46 antibody and (D) was taken with same Nikon Eclipse Ti camera

at 20x after pretreatment with anti-CD46/CD55/CD59 antibodies.

Page 57: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

45

2.4.5 Increasing complement challenge to BOEC results in an increase in

platelet adhesion

Atypical HUS is characterized clinically by thrombocytopenia, and pathologically by

the presence of platelet microthrombi.(Noris and Remuzzi, 2009) In order to model an

aHUS/TMA phenotype ex vivo we employed an endothelialized, microfluidic system,

so that calcein-labeled platelets could be perfused across a confluent monolayer of

BOEC and visualized for adhesion, after the BOEC had been exposed to a

complement challenge with 50% NHS alone or after regulator blockade. Minimal

platelet adhesion was demonstrated after the cells were exposed to complement active

serum for one hour or if the function of just CD46/MCP was blocked. However,

significant platelet adhesion occurred within 5-10 minutes of flowing the platelets

through the microfluidic channels when we augmented the complement challenge by

blocking the function of three surface complement regulators concurrently (p=0.028)

(Figure 2-5).

Page 58: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

46

Figure 2-5. Increasing complement challenge to BOEC results in a stepwise increase in

platelet adhesion

(A-B) Washed and calcein-labeled platelets in Tyrodes buffer (15 x 107/ml, 100μl/well) were

introduced into a microfluidic flow chamber and perfused at 2 dyne/cm2 from inlet to outlet

well. BOEC were pretreated with respective blocking antibody and 50% NHS in AP buffer

for 1 hour. Images were taken with a Nikon Eclipse Ti camera at 4x magnification after 5

minutes. A white line was added to better visualize upper and lower channel. In figure 5A the

upper chamber represents platelet adhesion after pretreatment with 50% NHS only, the lower

channel after pretreatment with CD46/MCP blocking antibody and 50% NHS. Figure 5B

shows in upper channel platelet adhesion after pretreatment with CD46/MCP blocking

antibody and 50% NHS and in lower channel adhesion after pretreatment with

CD46/CD55/CD59 blocking antibodies and 50% NHS for 1 hour. (C) Per viewing chamber,

three random pictures after 5 minutes of perfusion were taken and adhering platelets were

counted using ImageJ software. Number of platelets was normalized to number counted when

channels were perfused with media. N=6. * p<0.05 (paired t-test) (D) Image was taken in

microfluidic chamber using a Nikon Eclipse Ti microscope at 60x magnification oil

Page 59: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

47

immersion after BOEC were pretreated with CD46/55/59 antibodies, followed for 1 hour with

50% NHS and 10 minutes of platelets in Tyrodes buffer. Cells were fixed with 4% PFA/PBS,

blocked with 1% BSA/0.2% Triton X-100/PBS and stained for C5b-9 (rabbit polyclonal

antibody, 1:1000, red) and VWF (sheep polyclonal antibody, 1:1000, green) overnight. Alexa

fluor 488 and 555 (1:1000 for 1 hour) was used as secondary antibodies, Hoechst (0.12μg/ml,

10 minutes, blue) for DNA stain. Arrow indicates adhering platelets.

Page 60: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

48

2.4.6 Discussion

CD46/MCP acts as a cofactor for the serine protease CFI to inactivate cell surface

bound C3b. It is one of three principal membrane-anchored complement regulators

present on the surface of endothelial cells but the only one associated with aHUS.

Mutations in CD46/MCP, similar to other mutations that cause aHUS such as CFH

and CFI, are associated with low penetrance, but MCP/CD46-associated aHUS

generally has a more favorable outcome.(Caprioli et al., 2006) A mutation in

CD46/MCP may be the sole mutation demonstrated in patients with aHUS, or

MCP/CD46 variants may also represent a risk polymorphism, increasing the chance

of disease manifestation in a patient with another genetic susceptibility. (Bresin et al.,

2013; Provaznikova et al., 2012) Explaining how a mutation in CD46/MCP actually

leads to a TMA phenotype has not been clearly delineated.

In this study we found that a complete functional blockade of CD46/MCP, using a

monoclonal antibody directed towards its C3b binding site, was not followed by a

significant increase in cell surface C3 deposition, cell death, or platelet adhesion in an

ex vivo endothelial cell-based model system. In order to achieve the purported

sequence of events that lead to a TMA with complement deposition, endothelial cell

activation or death, and platelet adhesion, (Noris and Remuzzi, 2009) we had to

amplify the complement challenge by the synchronous/concomitant blockade of three

surface regulators. Only then, could we demonstrate a measurable increase in cell

surface C3b deposition, platelet adhesion and cell death. This implies that the

endothelial cell is well protected against complement injury and cumulative hits –

either endogenous or exogenous in nature – may be necessary to overcome this

protective barrier.

Experimental evidence of synergy between CD46/MCP and CD55/DAF already

exists, (Liszewski et al., 2007) and clinically, the presence of a risk haplotype in both

CD46/MCP and CFH doubles the penetrance as opposed to having just one, or neither,

and the combination doubles the risk of end stage renal disease. (Bresin et al., 2013)

Of course there may be an inciting „trigger‟, for instance, in one series, 100% of those

with aHUS and an CD46/MCP mutation had so in association with an infectious

Page 61: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

49

trigger. (Caprioli et al., 2006)

This study is the first study using blood outgrowth endothelial cells to study the

pathogenesis of a TMA - in this case CD46/MCP associated aHUS. BOEC are

thought to be a true EPC that possess distinctly endothelial cell characteristics and

behaviors. (Medina et al., 2010b; Tura et al., 2013) They are highly proliferative and

as compared to HUVEC they retain their phenotype over passages and are more

resistant to cell death. (Bompais et al., 2004) Already studied in various disease

models, (Medina et al., 2012) BOEC have significantly enhanced the understanding

of von Willebrand Disease by taking advantage of the fact that these cells can be

grown from patients expressing mutant endothelial cell proteins. (Starke et al., 2013;

Wang et al., 2013a) BOEC express the surface complement regulators similarly to

HUVEC, as we have shown, however in contrast to HUVEC, are highly proliferative

and stable over passages. Phenotypically they resemble more closely microvascular

endothelial cells, (Gremmels et al., 2011; Jiang et al., 2007; Toshner et al., 2014)

which make them an ideal cell type to study TMA pathogenesis, a disease of the

microvascular endothelium.

CD46/MCP mutations have been studied functionally, by either quantifying the

expression levels on peripheral blood mononuclear cells, or via transfection of CHO

cells, cells devoid of any complement regulators. (Caprioli et al., 2006) Studies of

transfected Chinese hamster ovary (CHO) cells have found that this reduced

expression translates into reduced regulation of the complement AP with increased C3

deposition and this is then thought to lead to enhanced formation and insertion of the

Membrane Attack Complex/ C5b-9 (MAC/C5b-9) into cells with endothelial cell

activation and/or death. (Liszewski et al., 2007) Furthermore, when CD46/MCP

expression levels were reduced by 50% on CHO cells, thus mimicking the situation of

a heterozygous mutation commonly associated with CD46/MCP-associated aHUS,

less efficient complement regulation and more C3b deposition was also found.

(Liszewski et al., 2007) Why we did not find more C3b deposition in the face of

efficient blockade of MCP and complement activation may speak to a synergistic

defensive effect of the other regulators.

The importance of conducting functional studies to characterize any given mutation as

Page 62: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

50

disease causing, even if the mutation is predicted to be pathogenic, has recently been

emphasized for aHUS associated with complement Factor B mutations. (Marinozzi et

al., 2014) Even then, explaining how a mutation leads to disease may not be clearly

evident. We believe that using our system incorporating BOEC and an endothelialized

microfluidic chamber that we are better able to model the in vivo scenario.

Our study has limitations of course, and the precise role of the other complement

regulators such as CFH was not studied. Platelet adhesion was assessed after one hour

of complement challenge and perhaps with more prolonged complement challenge

then a difference in CD46/MCP-blocked cells might have emerged, however time was

limited by having the cells outside of the 5% CO2 incubator.

In conclusion, our study suggests that the effect of a loss of CD46/MCP alone, on an

otherwise normal cell, endowed with the full complement of regulators, is probably

minimal. Our data supports the notion of a „multiple hit‟ – that an additional genetic

abnormality in the complement system (here simulated by blocking three surface

regulators) is needed for an individual with a CD46/MCP mutation to manifest the

disease. BOEC represent a novel tool to study aHUS pathogenesis being more

physiologically relevant than CHO cells, in that they express the complement

regulators, but more importantly, because they can now be isolated from patients with

mutations in CD46/MCP, enabling the study, ex vivo, of the „real‟ protein phenotype.

Page 63: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

51

2.4.7 Supplemental Data

Figure 2-S1. Functional blocking antibody binding and saturation

(A-C) Using flow cytometry the antibody saturation of anti-MCP antibody (GB24, A), anti-

CD55/DAF antibody (BRIC216, B) and anti-CD59 antibody (BRIC229, C) was determined.

Antibody concentrations of 2μg/ml (blue), 5μg/ml (orange) and 10μg/ml (green) were

compared to unstained control (red). Subsequently, 5μg/ml was used in all experiments. (D-F)

Co-localization of blocking antibody with epitope was determined using immunofluorescence.

The blocking antibodies were detected with secondary donkey anti mouse labeled with A555

(red) and staining antibody with representative Alexa fluor 488 (green). Hoechst dye (0.12

μg/ml, blue) was used to stain DNA. Pictures were taken with a spinning disk confocal

microscope as described in Methods and Figure legend 2-1.

Page 64: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

52

Figure 2-S2. Expression of surface-bound regulators on glomerular endothelial cells

compared to BOEC

(A-C) The expression of surface-bound complement regulators CD46/MCP, CD55/DAF and

CD59 on glomerular endothelial cells (GEC, green) and BOEC (purple) was determined

using flow cytometry. A similar expression pattern was observed for CD46/MCP (A),

CD55/DAF (B) and CD59 (C). A curve in grey represents unstained controls. N=1 (D)

mRNA expression of complement regulators were determined in control BOEC and GEC

using quantitative real-time PCR. Significant increased mRNA levels for GEC were

determined. N=3 in one experiment. * p<0.05, **** p<0.001 (two-tailed ANOVA).

Page 65: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

53

CHAPTER 3 VON WILLEBRAND FACTOR

MULTIMERS CONTRIBUTE TO COMPLEMENT

ALTERNATIVE PATHWAY CONTROL

Page 66: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

54

3.1 Abstract

Recent in vitro evidence supports a link between the alternative pathway of

complement (AP) and VWF based on the co-localization of complement activation

proteins and VWF multimers. It has been suggested that this link could possibly

amplify EC complement activation and therefore might have significant implications

for diseases where the crossover between complement and coagulation is evident,

especially the thrombotic microangiopathies such as atypical hemolytic uremic

syndrome (aHUS) and thrombotic thrombocytopenic purpura (TTP). Clinical

observations have shown a link between aHUS and TTP, as the phenotypes of both

entities significantly overlap. In addition, aHUS patients show variants in

ADAMTS13 and TTP patients show elevated complement activation parameters in

the acute phase of the disease. We aimed to use blood outgrowth endothelial cells

(BOEC), EC precursors that can be isolated from blood of patients, to further study

the role of VWF in the pathogenesis of aHUS and the link between the AP of

complement and VWF.

BOEC were cultured by a standard protocol from healthy controls and two patients

with type 3 von Willebrand disease patients (VWD3 BOEC), which have no VWF

secretion. EC phenotype and presence of complement regulators (CD46/MCP,

CD55/DAF, CD59) were confirmed by immunofluorescence (IF), western blot (WB),

qPCR and flow cytometry (FACS). Cells were challenged with complement using

50% normal human serum (NHS), with or without functional blockade of

complement regulators. BOEC death was assayed by LDH release.

BOEC isolated from healthy adult controls and two patients with type 3, von

Willebrand disease (VWD3), show similar surface expression of the tested

membrane-bound complement regulators. VWD3 BOEC are distinguishable for

having no visible Weibel-Palade bodies or VWF on immunofluorescence. VWF

secretion was confirmed from control BOEC after complement activation.

Immunofluorescently labelled complement products, including C5b-9, were

visualized in association with/in proximity to VWF released from complement

challenged control BOEC by spinning disk confocal microscopy. However,

complement (C3b) deposition was significantly greater on VWD3 BOEC following

complement challenge and these cells succumb more easily to complement-mediated

cytotoxicity. Taken together we have confirmed an association of VWF and

Page 67: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

55

complement AP activation products and the finding of increased complement

activation on BOEC devoid of VWF argues for a protective effect of this association.

Page 68: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

56

3.2 Introduction

Atypical hemolytic uremic syndrome (aHUS) and thrombotic thrombocytopenic

purpura (TTP) represent two diseases that present a thrombotic microangiopathy

(TMA) phenotype. Atypical HUS is a TMA that occurs due to dysfunctional

regulation of the complement system with microvascular endothelial cell injury and

the formation of platelet microthrombi. (Noris and Remuzzi, 2009) TTP is caused by

either a deficiency of a disintegrin and metalloproteinase with a thrombospondin type

1 motif member 13 (ADAMTS13) or autoantibodies that functionally inhibit its

function, allowing ultra large VWF multimers to accumulate, bind platelets and

produce occlusive thrombi. (Tsai, 2010)

Although TTP and aHUS have significant clinical overlap, making diagnosis a

challenge, they represent two distinct pathologies, both ultimately leading to a

microangiopathic hemolytic anemia. (Tsai, 2013) However, the line distinguishing

them may not be so clear-cut, and aHUS and TTP may well represent part of a

spectrum of complement-associated diseases. (Noris et al., 2012) The finding of a low

C3/hypocomplementemia in aHUS and TTP patients, (Noris et al., 1999) along with

the experimental observation that serum from TTP patients could result in

complement deposition on endothelial cells, (Ruiz-Torres et al., 2005) and the finding

of increased complement alternative pathway (AP) activity in TTP patients, (Feng et

al., 2013b) seems to confirm an overlap between the two entities. Furthermore, a

recent study found that 80% of an aHUS patient cohort also carried at least 1 non-

synonymous change in ADAMTS13, and in 38% of patients, multiple ADAMTS13

variations were found. These ADAMTS13 variants were thought to be clinically

relevant as measured ADAMTS13 activity was less than 60% in half of the patients

studied. (Feng et al., 2013a) This also argues in favor of an interconnection between

the two diseases. (Feng et al., 2013a)

Over the last year there have been a number of experimental studies further linking

the alternative complement pathway (AP) and its principal regulator, complement

Factor H (CFH), with von Willebrand Factor (VWF) and its cleaving protease,

ADAMTS13. (Feng et al., 2013c; Nolasco et al., 2013; Rayes et al., 2014; Turner and

Page 69: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

57

Moake, 2013) CFH has been shown to be a reductase for large soluble VWF

(LsVWF) multimers that are secreted from ECs.(Nolasco et al., 2013) LsVWF

multimers released from complement activated ECs assume a conformation that

makes them inaccessible to ADAMTS13 cleavage, but can still bind platelets and

induce platelet aggregation under conditions of high-fluid shear stress. (Nolasco et al.,

2013) In addition, the C-terminus of CFH has been shown to enhance ADAMTS13

VWF cleavage in vitro. (Feng et al., 2013c) Most interesting perhaps has been the

finding that both the assembly, and activation, of complement AP components on

ULVWF strings secreted from, and anchored to, endothelial cells (ECs). (Turner and

Moake, 2013) We aimed to explore this link further using blood outgrowth

endothelial cells (BOEC) lacking functional VWF, derived from patient with von

Willebrand type 3 disease.

Page 70: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

58

3.3 Methods

3.3.1 Establishing and characterizing blood outgrowth endothelial cells

(BOEC)

BOEC were isolated from the blood as described before in chapter 2. Briefly, 48ml

blood was drawn into cell preparation tube (CPT) vacutainers (Becton Dickinson,

Franklin Lakes, USA) and mononuclear cell layer was obtained by centrifugation at

1600xg for 30 minutes. Cells were washed twice in 10% fetal bovine serum (FBS:

Sigma-Aldrich, St. Louis, USA) in phosphate buffered saline (PBS, WISENT, St

Bruno, Canada), resuspended in cEGM-2 media with supplements (Lonza,

Walkersville, USA: Cat no. 362753) and seeded on six-well tissue culture plate

(Becton Dickinson, Franklin Lakes, USA) coated with 0.05 mg/ml of rat tail collagen

type I in 0.02 M acetic acid (Becton Dickinson, Franklin Lakes, USA). BOEC were

identified after 1-2 weeks by their endothelial cobblestone morphology and their

endothelial cell phenotype was confirmed by flow cytometry. BOEC are positive for

the endothelial cell (EC) surface markers CD31 (PECAM-1) and CD144 (VE-

Cadherin) while negative for the hemangioblast marker CD45 and the immature

endothelial progenitor cell marker CD14. BOEC were grown from healthy adult

donors (Control BOEC, CTRL BOEC) and patients with von Willebrand Disease type

3 (VWD BOEC).

3.3.2 Glomerular Endothelial Cell Culture

Glomerular endothelial cells (GEC) were cultured on T75 flasks (Sarstedt, Nümbrech,

Germany) coated with collagen (0.05 mg/ml of rat tail collagen type I in 0.02 M

acetic acid (Becton Dickinson, Franklin Lakes, USA)) in endothelial cell medium

(ScienceCell Research Laboratories, Carlsbad, USA, 1001) supplemented with

Endothelial Cell Growth Supplement (ECGS, ScienceCell Research

Laboratories ,1052), 5% fetal bovine serum (FBS, Sigma-Aldrich, St. Louis, USA)

and 1% Antibiotic-Antimycotic (Gibco, Invitrogen, Life Technologies, Carlsbad,

USA; containing 10,000 units/mL of penicillin, 10,000 µg/mL of streptomycin, and

25 µg/mL of Fungizone® Antimycotic). Cells were kept at 37°C and in an

environment with 5% CO2. Passages 4-7 were used for experiments.

Page 71: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

59

3.3.3 Determining BOEC protein expression via Western blot

In order to determine VWF expression levels in control and VWD BOEC, we

performed a western blot analysis of BOEC lysates. BCA protein determination assay

was used prior to preparation of samples for western blot. BOEC lysates were

prepared in 4 x sample buffer (40% (v/v) glycerol, 8% (w/v) sodium dodecyl sulfate

(SDS), 0.26M Tris, pH 6.8, 0.05% (w/v) bromphenol blue) and heated at 95°C for 5

minutes. 50 μg of protein were loaded and separated by an 8% SDS-polyacrylamide

gel electrophoresis running at 200 V for 45 minutes (Power Source 250V, VWR

International, Radnor, USA). Protein gels were transferred onto a 0.45 μm

nitrocellulose membrane (Bio-Rad Laboratories, Hercules, USA) for 70 minutes at

100 V (VWR International, Radnor, USA). Membranes were blocked with 5% (w/v)

skim-milk in Tris-Buffered-Saline, pH 7.6, + 0.05% (v/v) Tween-20 (TBST) for one

hour at room temperature. Membranes were incubated with rabbit polyclonal

antibody to VWF (1:1000 dilution; Dako, Glostrup, Denmark) in 5% (w/v) skim-milk

in TBST overnight at 4°C with shaking. For loading controls, membranes were

incubated with mouse monoclonal antibody to β-actin (β-actin, BA3R, 1:10000

dilution; Pierce Biotechnology, Rockford, USA) in 5% (w/v) skim-milk in TBST for

1 hour at room temperature with shaking. Membranes were washed 3 times, with

TBST for 5 minutes, and incubated with secondary antibody in 5% (w/v) skim-milk

in TBST at room temperature for 1 hour with shaking. Membranes were washed 3

times with TBST for 10 minutes. Proteins were detected using Western Lighting™

Plus-ECL, Enhanced (PerkinElmer, Waltham, USA) and developed on Odyssey FC

Imaging System (chemiluminescence detection, LI-COR Biosciences, Lincoln, USA)

using Image Studio software (LI-COR Biosciences, Lincoln, USA). Quantification of

protein bands was performed using densitometry (ImageJ).

3.3.4 Quantifying BOEC and GEC Gene Expression via qRT-PCR

To quantify VWF mRNA expression we performed quantitative real time polymerase

chain reaction (qRT-PCR). Cells were seeded overnight in a 6 well plate (Falcon) to

confluence, washed once with Hank‟s balanced salt solution (HBSS: Gibco, Life

Technologies, Carlsbad, USA) before RNA was isolated using TRI Reagent (Sigma-

Aldrich, St. Louis, USA) according to manufactures instructions. RNA concentration

and integrity was verified using Nanodrop (NanoDrop 1000, Thermo Fisher Scientific,

Page 72: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

60

Waltham, USA), and reverse transcribed using ReadyScript™ cDNA Synthesis Mix

(Sigma-Aldrich, St. Louis, USA, RDRT). Samples (200 ng cDNA in diethyl

pyrocarbonate (DEPC, Sigma-Aldrich) treated water) were amplified by real time

polymerase chain reaction (PCR) using StepOne™ System from Life Technologies

(Carlsbad, USA). Amplified products were detected using KiCqStart™ SYBR®

Green qPCR ReadyMix™, with ROX™ (Sigma-Aldrich, St. Louis, USA) and

analyzed as follows: 2–(C

T – C

T GAPDH) – C

T Control BOEC)

.

The following oligonucleotide primers purchased from Sigma-Aldrich, Carlsbad,

USA were used:

GAPDH: forward, 5‟-ACAGTTGCCATGTAGACC-3‟; reverse, 5‟-

TTTTTGGTTGAGCACAGG-3‟.

VWF: forward, 5‟-TGTATCTAGAAACTGAGGCTG-3‟; reverse, 5‟-

CCTTCTTGGGTCATAAAGTC-3‟.

3.3.5 Characterization of membrane-bound regulators of BOEC

For determination of surface expression of the membrane-bound regulators on VWD

BOEC we performed both immunofluorescence labeled imaging and flow cytometry.

3.3.5.1 Immunofluorescence

BOEC at a concentration of 0.5 × 106 cells/ml were seeded onto collagen-coated

22x22-mm cover slips (VWR International, Radnor, USA) and incubated overnight

(37°C, 5% CO2). Samples were washed with ice-cold PBS followed by fixation with

4% (w/v) paraformaldehyde (Electron Microscopy Sciences, Fort Washington, USA)

in PBS and blocked with 1% (w/v) BSA/PBS for 60 minutes. Samples were stained

overnight with the primary antibodies made up in the 1% (w/v) BSA/PBS solution.

Samples were washed in PBS and incubated for one hour with respective species

specific donkey-anti secondary antibodies conjugated with Alexa Fluor® 488 or

Alexa Fluor 555 (1:1000 dilution, Invitrogen, Life Technologies, Carlsbad, USA).

Cell nuclei were stained using 0.12 μg/ml Hoechst 33342 stain (Thermo Fisher

Scientific, Waltham, USA) for ten minutes. Samples were washed in PBS and cover

slips were mounted with Dako Fluorescence Mounting Media (Dako Canada,

Burlington, Canada) for microscopy. Images were taken with a spinning disk confocal

microscopy equipped with an Olympus IX81 inverted fluorescence microscope using

a 60./1.35 oil immersion objective equipped with a Hamamatsu C9100-13 back-

Page 73: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

61

thinned EM-CCD camera and Yokogawa CSU X1 spinning disk confocal scan head

(with upgrade from Spectral Aurora Borealis, Richmond Hill, Canada). The unit is

equipped with 4 separate diode-pumped solid state laser lines (Spectral Applied

Research, Richmond Hill, Canada, 405 nm, 491 nm, 561 nm, and 642 nm) with

emission filters: 447 nm ± 60, 525 nm ± 50, 593 nm ± 40, 620 nm ± 60, 676 ± 29 and

700 nm ± 75, and 1.5X magnification lens (Spectral Applied Research). Confocal

images were taken with an Improvision Piezo Focus Drive. Z-stacks were taken at

0.25 μm. Images were then deconvolved by iterative restoration using Volocity

Software (PerkinElmer, Waltham, USA) with confidence limit set to 95% and

iteration limit set to 20.

The following antibodies were used for IF: sheep anti-human VWF (1:1000 dilution,

AbD Serotec, Oxford, UK), rabbit polyclonal to C5b-9 (1:1000 dilution, Abcam,

1:1000 dilution) to CD46/MCP (1:50 dilution; Santa Cruz Biotechnology, Dallas, TX,

USA), rat polyclonal antibody to CD59 (1:1000 dilution; AbD Serotec, Oxford, UK),

and goat polyclonal antibody to CD55 (5 μg/mL; International Blood Group

Reference Laboratory, NHS Blood and Transplant, Bristol, UK).

3.3.5.2 Flow cytometry

Cells were seeded overnight in a 6 well plate (Falcon, Becton Dickinson, Franklin

Lakes, USA) to confluence, washed 1x with HBSS and then trypsinized (0.05%

Trypsin/0.53 mM EDTA, WISENT, St Bruno, Canada). The primary antibodies

(rabbit anti-CD46/MCP, 1:50 dilution, Santa Cruz Biotechnology, Dallas, USA, sc-

9098; goat anti-CD55/DAF, 1:100 dilution, R&D Systems, Minneapolis, USA,

AF2009; rat anti-CD59, 1:100 dilution, AbD Serotec, Oxford, UK, MCA715G) and

secondary antibodies (Alexa fluor 488, 1:200 dilution, Invitrogen) were incubated at

4°C for 20 minutes. At least 10,000 events were recorded using Attune Acoustic

Focusing Cytometer (Invitrogen) and FlowJo vX.0.7 software (FlowJo LLC, Ashland,

USA). Results are given as median fluorescence intensity (MFI). Cells were gated for

live cells (red laser 536nm, emission channel 2), single cells (FSC-A vs. FSC-H) and

finally through forward scatter and side scatter to determine the BOEC population.

Surface regulators were recorded via the blue laser 488 nm, emission channel 1. To

Page 74: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

62

correct for spectral overlap during multicolor flow cytometry experiments, color

compensation was performed each time.

3.3.6 Complement Challenge of BOEC

In order to challenge the BOEC via the alternative pathway of complement, cells were

exposed to 50% normal human serum (NHS) with alternative pathway (AP) buffer (7

mM MgCl2, 10 mM EGTA, 144 mM NaCl, 20 mM HEPES buffer, pH 7.4). To

escalate / enhance the challenge to the cells, the membrane-anchored complement

regulators CD46/MCP, CD55/DAF and CD59 were functionally blocked, prior to the

addition of 50% NHS.

3.3.6.1 Normal human serum

50% normal human serum was used as source of complement. Serum was collected

from whole blood of adult donors into serum vacutainers (BD Biosciences), allowed

to clot for 30 minutes and then centrifuged at 3000 x g at 4°C. Serum was stored at -

20°C until needed as a source for complement. Heat-inactivated serum (HIS), for use

as a negative control, was obtained by incubating the serum for 30 minutes at 56°C.

3.3.6.2 Membrane-anchored complement regulator blockade

In order to simulate a CD46/MCP mutation, a non-complement-activating (Schiff et

al., 2004) monoclonal mouse anti-human anti-CD46/MCP antibody (GB24, IgG1,

kindly provided by John Atkinson, St Louis, MO, USA) was used. Additional

blockade of EC surface complement regulation was achieved with the use of a

monoclonal anti-human CD55/DAF (BRIC216, IgG1) and monoclonal anti-human

CD59 (BRIC229, IgG2b) (International Blood Group Reference Laboratory, NHS

Blood and Transplant, Bristol, UK) functionally blocking antibodies. Antibodies

were used at a concentration of 5 μg/mL and diluted in serum free cEGM-2 (Lonza)

media for 30 minutes in all experiments. The concentration of blocking antibody used

was derived from antibody titration and saturation experiments performed by FACS,

as detailed in chapter 2.

Page 75: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

63

3.3.7 Assessment of Complement Challenge of BOEC

3.3.7.1 Complement deposition detected by immunofluorescence

For IF demonstration of complement deposition (C3 and C5b-9), BOEC were grown

on collagen-coated coverslips (VWR) and then incubated for 60-240 minutes (37°C,

5% CO2) with either cEGM-2 (BOEC) media, 50% normal human serum (NHS) in

AP buffer alone or after complement regulator blockade. After 60-240 minutes the

supernatant was aspirated and the cells were washed 3 times with ice cold PBS,

blocked with 1% (w/v) BSA for 1 hour and incubated with rabbit polyclonal antibody

to C3c, (Abcam, Cambridge, UK, ab15980, 1:1000 dilution) or a rabbit polyclonal

antibody to C5b-9 (Abcam, Cambridge, UK ab55811, 1:1000 dilution) overnight.

3.3.7.2 Complement deposition detected by FACS

C3b deposition was demonstrated by FACS using an antibody to C3c (rabbit

polyclonal, Abcam) that detects C3c as well as the C3c part of native C3 and C3b.

Cells were seeded overnight in a 6 well plate (Falcon) to confluence, washed 1x with

HBSS before blocking antibodies (GB24, BRIC216, BRIC229) were added for 30

minutes in serum-free media, followed by 50% NHS in AP buffer for 1 hour. Cells

were trypsinized (0.05% Trypsin/0.53 mM EDTA, WISENT) and washed in Flow

buffer (FB, 1% FBS/PBS) twice. The primary antibody (rabbit anti-C3c, 1:100

dilution, Abcam, ab15980,) in FB and the secondary antibody (R-Phycoerythrin-

conjugated AffiniPure F (ab‟) 2 Fragment Donkey Anti-Rabbit IgG (H + L), 1:200

dilution, Jackson ImmunoResearch, West Grove, USA) together with Fixable

Viability Dye eFluor780 (1:1000 dilution, eBioscience, San Diego, USA, 65-0865) in

PBS were incubated at 4° for 20 minutes. Cells were analyzed using Attune Acoustic

Focusing Cytometer and FlowJo 10.X.0 software. Results are given as median

fluorescence intensity (MFI). Cells were gated for live cells (red laser 536nm,

emission channel 2), single cells (FSC-A vs. FSC-H) and finally through forward

scatter and side scatter to determine the BOEC population. C3 deposition was

recorded via the blue laser 488nm, emission channel 2. To correct for spectral overlap

during multicolor flow cytometry experiments, color compensation was performed

each time.

Page 76: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

64

3.3.8 Assessing the impact of complement challenge

3.3.8.1 LDH cell cytotoxicity assay

The LDH cell cytotoxicity assay was used to quantify cell death and compare the

impact of complement challenge on the different cell lines. BOEC from three healthy

controls and from two VWD type 3 patients were seeded overnight in a 96 well

ELISA plate (Sarstedt, Nümbrech, Germany) and grown to confluence. Prior to the

experiment cells were washed once with HBSS before, if indicated, adding the

complement-blocking antibodies diluted in serum-free media (cEGM-2) for 30

minutes. After washing twice, 10% NHS in serum-free media was added for 4 hours.

Pierce LDH cytotoxicity assay kit (Thermo Fisher Scientific, Waltham, USA) was

used according to manufacturer‟s instructions. Optical density (OD) was calculated

using a standard curve and then normalized to positive control and displayed in

percent increase over negative control (10%NHS). The positive control represents

maximal release, which was achieved by solubilizing cells with lysis buffer.

3.3.8.1 Platelet adhesion Assay

Whole blood was collected from healthy adult donors with acid citrate dextrose

(ACD: 22.9 mM citric acid, 44.9 mM sodium citrate dehydrate, 74 mM dextrose

monohydrate) anticoagulation. Platelet rich plasma (PRP) was attained by

centrifugation of the whole blood at 160 x g for ten minutes. The PRP was spun at

950 x g for seven minutes to pellet platelets which were then washed twice with

PBS/ACD solution (20% ACD in PBS; pH 6.1). Platelet count was measured using an

automatic hematocytometer (Beckman Coulter, Brea, USA) and concentration was

adjusted accordingly with PBS/ACD. Platelets were incubated with 2.5 μM calcein

(Life Technologies, Carlsbad, USA) for thirty minutes at 37°C. Finally, platelets were

pelleted at 950 x g and resuspended at a concentration of 15 x 107/ml in Tyrodes

buffer (136 mM NaCl, 2.7 mM KCl, 0.42 mM NaH2PO4, 19 mM NaHCO3, 5.5 mM

of glucose, 1 mM CaCl2, 1 mM MgCl2 and 10 mM of hyroxyethyl

piperazineethanesulfonic acid (HEPES: Invitrogen, Life Technologies, Carlsbad,

USA).

For BOEC-platelet adhesion experiments, control BOEC and VWD type 3 BOEC

(passage 3-14) were grown in collagen-coated microfluidic channels of the BioFlux

Page 77: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

65

system (Fluxion Biosciences, South San Francisco, USA) as described before (in

chapter 2). Cells were washed at 1 dyne/cm2 with HBSS (Invitrogen). For

complement challenge experiments the membrane complement regulators were first

blocked with anti-human CD46/MCP, CD55/DAF and CD59 functional blocking

antibodies diluted in serum-free media (cEGM-2) and perfused through the channels

at 1-2 dynes/cm2 for thirty minutes. 50% NHS from healthy adult donors in

alternative pathway (AP) buffer was subsequently perfused through the BioFlux

chambers at a shear rate of 2 dynes/cm2 for 60 minutes. For platelet adhesion assays,

15 x 107/ml calcein labeled platelets in Tyrodes buffer (as described in platelet

preparation section) were flowed through the chamber at 2 dyne/cm2 for 10 minutes

after BOEC exposure to serum. Per time point and experiment three pictures of each

channel ([4x] magnification, NIKON camera) were saved and platelet adhesion was

manually counted using ImageJ software.

3.3.9 Ethics

Ethics approval was obtained from the Research Ethics Board at The Hospital for

Sick Children, Toronto, ON, Canada.

3.3.10 Statistical analysis

Figures were generated using GraphPad Prism 6 and displayed as mean and standard

deviation. Statistical analysis was performed using either two-way ANOVA with

post-hoc analysis or paired t-test. A p value < 0.05 was considered as statistically

significant. In figures p values are presented as follows: * ≤0.05, ** ≤0.01, ***≤0.001,

****≤0.0001, ns > 0.05.

Page 78: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

66

3.4 Results

3.4.1 BOEC Possess Endothelial Cell Characteristics

Blood outgrowth endothelial cells are endothelial progenitor cells. BOECs were

isolated from the peripheral blood of healthy adult volunteers (control BOEC) and

from two patients with von Willebrand disease type 3 (VWD BOEC). The latter lack

functional von Willebrand factor (VWF antigen levels were undetectable) due to

compound heterozygous mutations (c. 876delC, c. 1255C>T; c. 3939G>A, c. 5842+1

G>C) in both patients. Firstly, an endothelial phenotype was confirmed by

immunofluorescence and flow cytometry. The presence of Weibel-Palade bodies

(WPB) containing VWF in the control BOEC confirmed an endothelial phenotype.

WPB are present only in endothelial cells and megakaryocytes. An endothelial cell

phenotype of both control and VWD BOEC was corroborated by demonstrating the

presence of key endothelial cell markers by flow cytometry (positive for CD144/ VE-

Cadherin and CD31/Platelet endothelial cell adhesion molecule (PECAM-1), as well

as ensuring that the cells were not of an immature progenitor cell or of a

hemangioblast origin (negative for CD45 and CD14). This has been previously

described in more detail in Chapter 2.

Next we confirmed by western blot and qRT-PCR the minimal expression of VWF in

BOECs isolated from the two patients with type 3 VWD. VWD BOEC show only

5.6% of the mRNA levels of control BOEC (Figure 3-1). Of note, VWF mRNA levels

in glomerular endothelial cells were also decreased compared to BOEC (35% of

mRNA level in control BOEC, p< 0.0007, paired t-test, Supplement 3-1).

Page 79: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

67

Figure 3-1. BOEC possess endothelial cell characteristics

(A) Lysates of control BOEC (CTRL) and VWD BOEC (VWD) were probed for von

Willebrand factor (VWF, 1:1000) on an 8% SDS-page. VWF multimers were seen in CTRL

BOEC, whereas a significantly reduced amount of VWF was seen in VWD type 3 BOEC. (B)

This was confirmed by qPCR, where VWD BOEC showed just a minimal mRNA expression

of VWF (5.6% of control, p<0.0001, paired t-test). VWF mRNA levels were normalized to

GAPDH and control BOEC.

3.4.2 Type 3 VWD BOEC Express Similar Amounts of Membrane-

Anchored Complement Regulators as Control BOEC

In order to further characterize the VWD3 BOEC as relevant to study aHUS

pathogenesis we confirmed the presence of the membrane-anchored complement

regulators CD46/MCP, CD55/DAF and CD59 by immunofluorescent labeling and

visualization by spinning disk confocal microscopy (Figure 3-2 A-C). We next

demonstrated using flow cytometry that BOEC isolated from patients with type 3

VWD had quantitatively similar expression levels of the complement regulators

CD46/MCP, CD55/DAF and CD59 on their surface (Figure 3-2 D-G). VWD BOEC

had a 4.5-fold higher level of CD55/DAF and a 35-fold higher level of CD59 as

compared to CD46, exhibiting an equivalent cell surface distribution of the regulators

as compared to control BOEC (Figure 3-2 G).

Page 80: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

68

Figure 3-2. Type 3 VWD BOEC express similar amounts of membrane-anchored

complement regulators as control BOEC

By immunofluorescence (A-C) and flow cytometry (D-G) the surface expression of

complement regulators CD46/MCP, CD55/DAF and CD59 was detected. (A-C) Cells were

seeded on cover-slips, stained for CD46/MCP (1:50; A), CD55/DAF (1:200; B) and CD59

(1:1000; C) and the representative secondary antibody (Alexa fluor 488, green) and imaged

using a Olympus IX81 inverted fluorescence microscope with a 60/1.35 oil immersion

objective and a Hamamatsu C9100-13 back-thinned EM-CCD camera with Yokogawa CSU

X1 spinning disk confocal scan head. Confocal images were taken with an Improvision Piezo

Focus Drive. Z-stacks were taken at 0.25 μm. Images were deconvolved by iterative

Page 81: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

69

restoration using Volocity Software with confidence limit set to 95% and iteration limit set to

20. Cell nuclei were stained using 0.12 μg/ml Hoechst 33342 stain (blue).

(D-G) For flow cytometry cells were trypsinized off a 6-well plate, and incubated with

primary antibody (CD46, 1:50; CD55, 1:100; CD59; 1:100) and respective secondary Alexa

fluor 488 (1:200). Surface expression of complement regulators was acquired using Attune

Acoustic Focusing Cytometer (Invitrogen) and analyzed using FlowJo software. A similar

surface expression of complement regulators on VWD BOEC (blue) compared to control

BOEC (red) was observed, as shown in representative images D-F. The unstained controls are

displayed in dark grey for VWD BOEC and light grey for control BOEC. (G) Comparison of

the median fluorescence intensity (MFI) of three experiments did not show a significantly

different surface expression of CD46/MCP, CD55/DAF and CD59 (p=0.69, two-way

ANOVA).

3.4.3 Complement activation products associate with VWF

Release of VWF from endothelial cells is known to occur in response to complement

activation. This is dependent on the insertion of the terminal complement complex

C5b-9 in the cell membrane and seemingly independent of the anaphylatoxins C3a or

C5a. (Hattori et al., 1989) Recently the association of complement activation products

with endothelial cell derived VWF, has been shown in both static and fluidic

conditions.(Tati et al., 2013; Turner and Moake, 2013) As a first step we stained

concomitantly for both C5b-9 and VWF by immunofluorescence labeling of fixed

BOEC that had been exposed to 50% NHS in AP buffer. This was performed in static

conditions where BOEC, grown to confluence on coverslips, were incubated for one

hour with 50% NHS. By IF, the presence of positive C5b-9 staining on cells was

associated with VWF released from the cell. Intensification of complement challenge,

achieved by blocking three membrane-anchored regulators (CD46/MCP, CD55/DAF

and CD59) and exposing the BOEC to 50% NHS for either one or two hours was

associated with a reduction in the number of visible VWF-positive WPB. Not all cells

demonstrated C5b-9 deposition and in these cells the WPB were still clearly visible.

This was confirmed also under fluidic conditions where the BOEC were grown to

confluence in the BioFlux microfluidic chamber system prior to exposure to 50%

NHS in AP buffer. Representative pictures of experiments repeated on at least 4-6

separate occasions with 3-4 separate BOEC cell lines are shown in figure 3. Finally,

Page 82: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

70

we demonstrated an apparent co-localization of C5b-9 with VWF multimers (Figure

3-3).

Figure 3-3. Complement challenge results in VWF release and the association of

complement activation products and VWF

(A-C) Control BOEC were exposed to 50% heat- inactivated serum (HIS) for 2 hours (A),

50% NHS after functional blockade of complement regulators CD46/MCP, CD55/DAF and

CD59 for 1h (B) and 2h (C). Incremental complement blockade results in VWF exocytosis

and subsequent C5b-9 deposition. (D-F) Exposing control BOEC to shear (2dyne/cm2) in a

Page 83: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

71

microfluidic (BioFlux) flow chamber and 50% NHS/AP buffer also resulted in VWF release

and the formation of VWF strings (1:1000, green). (B) C5b-9 deposition was detected using a

rabbit polyclonal antibody (1:1000, red). (C) The merge indicates a co-localization of C5b-9

on VWF strings. Pictures were taken with a Nikon Eclipse Ti camera, 60x magnification/oil

immersion.

3.4.4 Platelet adhesion in response to complement challenge is initially

dependent on VWF release

In the complement-mediated aHUS form of TMA, dysregulation of the complement

alternative pathway (AP) is thought to ultimately lead to endothelial cell activation, a

procoagulant endothelium and platelet aggregation. Whether the platelets adhere to

VWF, injured cells or exposed subendothelial matrix or all of these and at what

timepoint this happens has not been clearly delineated. To investigate the functional

role of VWF in this process we sought to measure platelet adhesion to complement-

challenged BOEC grown in a microfluidic flow chamber while comparing normal

BOEC with those from type 3 VWD patients that lack VWF. In pursuance of this aim,

control and type 3 VWD BOEC were seeded overnight in the flow chamber and

washed, calcein-labeled platelets (15x107/ml) were perfused at 2 dyne/cm

2 for 10-20

minutes. Platelet adhesion on VWF strings (Figure 3-4 B-C) was achieved when

BOEC were treated with only 50% NHS and even more after functional blocking of

surface-bound regulators CD46/MCP, CD55/DAF and CD59 (20 platelets/hpf versus

84 platelets/hpf at 4X magnification). Platelets can be seen to adhere in control

BOEC only and not to VWD BOEC devoid of VWF (p<0.01). (Figure 3-4).

Page 84: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

72

Figure 3-4. Platelet adhesion in response to complement challenge is initially dependent

on VWF release

Control and VWD BOEC were seeded overnight in microfluidic (BioFlux) 48-well chamber.

If needed cells were blocked with CD46 or CD46/55/59 blocking antibodies for 30 minutes at

1 dyne/cm2 prior to perfusion with 50% NHS in AP buffer for 1 hour. Washed, calcein-

labeled platelets (2.5μM) were introduced from inlet to outlet well at 2 dyne/cm2. Pictures

were taken with a Nikon Eclipse Ti camera at 4x (A-B) and 20x (C) magnification after 5

minutes. (A) Channels containing VWD BOEC (upper channel) and control BOEC (lower

channel) were exposed to 50% NHS/AP buffer for 1 hour and platelets (15 x107/ml,

100μl/well) in Tyrodes buffer were perfused at 2 dyne/cm2 for 5 minutes before pictures were

taken (4x magnification). Minimal platelet adhesion occurred on control BOEC. (B) With

increasing complement challenge, induced by incremental blockade of complement regulators

CD46/MCP, CD55/DAF and CD59, platelet adhesion only occurred when VWF strings were

secreted by control BOEC (4x magnification). (C) Image of control BOEC (treated as in B)

clearly shows platelet adhesion to VWF strings (20x magnification). (D) Platelet adhesion

was analyzed by counting adherent platelets of four random pictures/channel using ImageJ

software. Significantly more platelets can be seen to adhere in control BOEC as

compared to VWD BOEC devoid of VWF (**

p<0.01, two-tailed ANOVA, Sidak's

multiple comparisons test). N=3

Page 85: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

73

3.4.5 VWD BOEC Show Increased C3b Deposition after Complement

Challenge

To investigate whether VWF has an amplifying role for the complement AP on the

endothelium we performed flow cytometry for C3b deposition on control and VWD

BOEC under increasing complement challenge as follows; (i) 50% NHS/AP buffer

alone, (ii) the functional blockade of one complement regulator (CD46/MCP)

followed by 50% NHS/AP buffer and (iii) the functional blockade of three surface

regulators (CD46/MCP, CD55/DAF and CD59). We observed an increase of the

median fluorescence intensity (MFI) in VWD BOEC treated with 50% NHS after

functional blockade of complement regulators compared to control BOECs (MFI

5861±3332 vs. 3450±1599, p<0.05, N=5, Figure 3-5).

Figure 3-5. VWD BOEC show increased C3b deposition after complement challenge

Control and VWD BOEC were seeded overnight in 6-well plates and treated with functional

blocking antibody and 50% NHS in AP buffer. Cells were washed, trypsinized and incubated

with primary antibody polyclonal rabbit anti-C3c (1:100), secondary donkey anti rabbit R-

Phycoerythrin (1:200) and fixable viability dye (eFluor 780, 1:1000) at 4° for 20 minutes.

Surface expression of C3b was acquired using Attune Acoustic Focusing Cytometer

(Invitrogen), after gating for live and single cells. Experiments were performed using two

different control BOEC and two different VWD BOEC. (A) shows one representative figure:

unstained control BOEC (light green), unstained VWD BOEC (dark green), 50% NHS

control BOEC (red), 50% NHS VWD BOEC (orange), CD46/55/59 block + 50% NHS

control BOEC (blue) and CD46/55/59 block + 50% NHS VWD BOEC (purple). (B) The

Page 86: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

74

Median Fluorescence intensity (MFI) was calculated using FlowJo software after subtracting

MFI of unstained sample.

3.4.6 VWD BOEC Are More Vulnerable To Complement-Mediated

Cytotoxicity

We next wanted to confirm that this increase in complement deposition seen by flow

cytometry translated into an increase in complement-mediated cell injury and death.

Cell cytotoxicity was quantified by measurement of LDH released from the

complement-challenged BOEC into the supernatant. Cell death of VWD BOEC was

increased as compared to control BOEC with mild (50% NHS alone), moderate (50%

NHS after CD46/MCP blockade) or severe (50% NHS after CD46/CD55/CD59

blockade) complement challenge. The adjuvant block of three surface-bound

complement regulators achieved a cumulative effect. VWD BOEC demonstrated a

46±14% rate of cell death when treated with 10% NHS, a 49±16% when treated with

50% NHS after CD46 block and a 81±20% when incubated with 50% NHS after

block of CD46/MCP, CD55/DAF and CD59. This is compared to a rate of cell death

of 31±15%, 30±12% and 48±28%, respectively in control BOEC (p<0.01, p<0.05,

p<0.05, Figure 3-6).

Page 87: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

75

Figure 3-6. VWD BOEC are more vulnerable to complement-mediated cytotoxicity

Cell death was measured by detection of LDH release in supernatant of control and VWD

BOEC after incubation with 10% NHS in serum-free media for 4 hours after pretreatment

with either none or CD46 or CD46/55/59 blocking antibodies. Cell death was calculated using

a standard curve and normalized to positive control (100%), obtained adding lysis buffer 45

minutes prior to incubation end. Background cell death from control BOEC was subtracted

for each value. Data was gathered from three different experiments (mean of 4-8 wells/plate)

using two different control BOEC and two different VWD BOEC. A more profound increase

of cell cytotoxicity was observed in VWD BOEC compared to control BOEC in all conditions.

* p<0.05 **p<0.01, ***p<0.001 (paired t-test).

Page 88: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

76

3.5 Discussion

We aimed to study the interaction between complement and VWF in TMA

pathogenesis. Using BOEC isolated from type 3 von Willebrand disease patients we

were able to take advantage of a naturally occurring endothelial cell line with

negligent amounts of VWF, essentially equating to a VWF knockout, to study the role

played by VWF in the pathogenesis of a TMA initiated by complement dysregulation

and the association of VWF and the alternative pathway of complement. It is known

that complement activation leads to EC activation and VWF release. (Hattori et al.,

1989; Nakashima et al., 2002; Ota et al., 2005) The first question we addressed was

how important this exocytosed VWF is for platelet adhesion in the setting of an acute

complement stress to the EC. In a microfluidic flow chamber (BioFlux) we were able

to see that platelet adhesion after complement-induced EC activation was primarily

VWF dependent, as no platelet adhesion was observed in VWD BOEC.

Multimerized VWF dimers are compactly stored in the WPB of endothelial cells.

Exocytosed VWF multimers can remain attached to EC and play a pivotal role in the

initiation of hemostasis by providing a platform on which platelets can

adhere.(Valentijn et al., 2011) When released from EC these multimers can form

(bundles of) strings which unfold in the circulation under the influence of shear flow

thus unveiling the glycoprotein (GP) 1b platelet-binding site.(Nightingale and Cutler,

2013) Normally these strings are subject to proteolysis by ADAMTS13.

Alternatively, released VWF can bind to collagen. (Ruggeri, 2007) Secreted VWF

differs from that in the circulation, which is present in a globular state, thus

maintaining both the platelet GP1b binding site and the ADAMTS13 cleavage site

cryptic/hidden. (Nightingale and Cutler, 2013)

To further investigate the interaction of VWF and the AP of complement, we utilized

BOEC from type 3 von Willebrand disease patients. It was first reported that in static

conditions, complement AP products, including C3 and C5, were present on

endothelial cell secreted VWF strings. (Turner and Moake, 2013) Tati et al showed

that, under shear conditions and in the absence of ADAMTS13, C3 bound to

histamine-induced VWF strings, to VWF adherent platelets, and to the endothelial

cell that secreted the VWF. (Tati et al., 2013) However, the functional downstream

Page 89: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

77

effect of this binding remained unclear. (Tati et al., 2013) Expanding on these studies,

we found that when BOEC were subjected to an incrementally increased complement

challenge in a microfluidic chamber that C5b-9 deposition was associated with the

VWF multimers as well as being deposited on the endothelial cell.

This raised the question as to whether the association of the AP and VWF was

actually complement amplifying resulting in enhanced complement deposition on EC

– a theory that has also been recently proposed by others. (Turner et al., 2014)

Considering this hypothesis, we expected to find less complement activation on cells

devoid of VWF. However, after exposing VWD BOEC to a complement challenge

(50% NHS with and without regulator blockade) we demonstrated increased C3b

deposition on their surface as compared with control BOEC. As VWD BOEC

expressed similar amounts of complement regulators CD46/MCP, CD55/DAF and

CD59 on their surface we ruled out other reasons for this result. This was

biologically relevant, as VWF deficient BOEC exhibited decreased survival to

complement-mediated cytotoxicity. Taken together, this is highly suggestive of a

protective role for VWF release or it limits complement deposition on EC surface.

It also raises the question as to whether cells perhaps expressing or containing less

VWF are more vulnerable to complement mediated injury. The heterogeneity of the

vascular endothelium is diverse and known to include VWF expression. (Aird, 2007a,

b) In lung endothelium, for example, VWF expression is strongest in veins and very

weak in pulmonary capillaries. (Kawanami et al., 2000; Muller et al., 2002) The

fenestrated glomerular endothelium had patchy positivity for VWF as detected by

immunohistochemistry on biopsy and post mortem specimens in one study.

(Pusztaszeri et al., 2006) This could partially explain why the glomerular endothelium

is vulnerable to complement attack in aHUS and perhaps why in TTP the kidney is

classically spared. We confirmed that cultured GEC have less VWF by both PCR and

Western blot and on immunofluorescence it was apparent that most GEC completely

lack VWF. (Supplementary Figure 3-6)

Our findings suggest two phases in the pathogenesis of TMA and aHUS. Initially,

complement activation leads to EC activation and VWF release, which is a protective

response, to facilitate platelet adhesion and delivery to the site of microvascular EC

Page 90: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

78

injury and to sequester complement activation products. In later stages, with

prolonged complement challenge, EC injury may lead to cell death with tissue factor

release and platelet adhesion to the exposed basement membrane components or

apoptotic/dying cells. Loss of VWF from the EC into the circulation in response to

ongoing complement-mediated WPB exocytosis may leave the cells more vulnerable

to complement injury.

In conclusion, by comparing Type 3 VWD BOEC and control BOEC and their

response to complement challenge we have been able to demonstrate that VWF

multimers released by the vascular EC contributes to EC protection by acting as

negative complement regulator. Similarly, lack of VWF results in increased

complement deposition and cell cytotoxicity. Different from recent claims in the

literature, we here demonstrate for the first time a new principle in EC complement

regulation via VWF. Our results overcome the assumed dichotomy of complement

and VWF (or aHUS and TTP) as separately acting biological pathways and provide

evidence for an intimate functional link between the two systems. This insight not

only contributes to a better understanding of TMA pathogenesis but also points

towards more efficient strategies for the monitoring and treatment of TMA patients.

Page 91: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

79

3.6 Supplemental Data -VWF expression in glomerular

endothelial cells

Figure 3-S1. Glomerular endothelial cells express less VWF

(A, B) BOEC and glomerular endothelial cell (GEC) lysates were resolved by 8% SDS-

PAGE and probed for VWF (1:1000). A lower expression of VWF was seen in GEC on

western blot, (B) which was quantified using densitometry, and revealed a VWF expression

of 60% in GEC. N=1. (C) By qPCR the mRNA level of von Willebrand Factor (VWF) in

control BOEC and glomerular endothelial cells (GEC) was measured. GEC show 35% of

VWF mRNA levels than BOEC (*** p<0.001, two-tailed t test, N=3) (D)

Immunofluorescence labeling of GEC with antibodies detecting VWF (1:1000, red) and

CD59 (1:1000, green) revealed partial expression of VWF in glomerular endothelial cells.

Page 92: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

80

Image is shown in extended focus and was taken using a Olympus IX81 inverted fluorescence

microscope with a 60/1.35 oil immersion objective and a Hamamatsu C9100-13 back-thinned

EM-CCD camera with Yokogawa CSU X1 spinning disk confocal scan head. Confocal

images were taken with an Improvision Piezo Focus Drive.

Page 93: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

81

CHAPTER 4 UNIFYING DISCUSSION, FUTURE

DIRECTIONS AND CONCLUSIONS

Page 94: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

82

4.1 Unifying Discussion

TMA is a severe multisystem disease involving endothelial cell damage, thrombosis

of the microvasculature and organ dysfunction. Although significant advances have

occurred in understanding the pathophysiology underlying the expanding spectrum of

diseases associated with TMA, coupled with improvements in its diagnosis and

therapy, there are still significant gaps in knowledge, areas of uncertainty and patients

dying from this disease. (George et al., 2012; George and Nester, 2014) Advancing

the understanding of TMA pathology will inevitably lead to new and improved

treatment strategies and translate into better outcomes for TMA patients. In order to

achieve this goal, we developed a model system to study the pathogenesis of TMA

using an endothelial progenitor cell (BOEC). BOEC were examined under static and

microfluidic conditions and exposing them to a complement challenge, achieved by

incrementally impairing complement control by functionally blocking membrane

regulators. A set of reliable, reproducible and disease-relevant readouts was

established, including demonstration of complement deposition by FACS,

quantification of cell death, and platelet aggregation under microfluidic conditions.

Designed to more closely resemble the patients‟ genotype and phenotype, in the

future, we will utilize patient derived BOEC, serum and other constituents such as

platelets to thus achieve an ex vivo test system that resembles an individual patient‟s

pathology in an unprecedented way.

This system was then used to study CD46/MCP associated aHUS. Loss of just 50%

CD46/MCP function can be associated clinically with recurrent aHUS, while a person

with complete knockout of the gene may or may not manifest disease. (Couzi et al.,

2008) Studies performed in vitro have found these mutations to be disease relevant.

These studies have been performed in transfected CHO cells where these mutated

CD46/MCP forms are associated with failure to inactivate cell surface C3b deposited

on cells after complement challenge. Although the CHO system is a „clean‟ system,

allowing delineation of the specific effect of a given mutation, it does not exemplify

the in vivo situation, as these cells are naturally devoid of other regulators. In our

model system, using BOEC, a cell endowed with the other complement regulators,

completely blocking the function of CD46/MCP failed to lead to a measurable or

significant increase in complement deposition, endothelial cell death or platelet

Page 95: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

83

adhesion. Instead, it was necessary to augment the „complement challenge‟ by

blocking three of the membrane-anchored regulators to achieve the phenotype.

This finding firstly provides support for the hypothesis that additional (genetic)

aberrancies in the complement system could increase the penetrance of these

CD46/MCP mutations and even modulate severity of disease. (Bresin et al., 2013)

Additional genetic „hits‟ in complement genes and regulators have been described in

patients with CD46/MCP mutations. (Bienaime et al., 2010; Bresin et al., 2013;

Fremeaux-Bacchi et al., 2013; Geerdink et al., 2012; Provaznikova et al., 2012;

Stevenson et al., 2014) Secondly it highlights the importance of functional studies in

helping to understand how a given mutation may lead to disease, and the utility of

different model systems. This has been recently emphasized and is especially relevant

considering the extent to which predictive software is now used to suggest the

likelihood of disease relevance or pathogenicity of a given mutation. (Marinozzi et al.,

2014)

The two archetypal representations of TMA, namely TTP and HUS, have many

shared elements in terms of clinical symptoms and pathology. More recently

complement has emerged as a common bond between the two. (Noris et al., 2012; Wu

et al., 2013) Expanding on this theme, and considering the recent data linking VWF

and the AP of complement (Turner and Moake, 2013) as well as the postulated

complement activating or amplifying role for that interaction (Turner et al., 2014) we

set about using BOEC naturally deficient of VWF to confirm this. Different from this

prediction, however, complement activation was actually greater on these cells rather

than less and the cells were more vulnerable to complement mediated cytotoxicity.

This suggests a regulatory role for VWF multimers released from EC upon

complement-stimulation.

Considering that glomerular endothelial cells have been shown to have very minimal

VWF, (Pusztaszeri et al., 2006) this finding may explain why glomerular endothelial

cells are more prone to complement-mediated injury, thus explaining the

susceptibility of the kidney to complement-mediated TMA. Furthermore, using the

BOEC lacking VWF and comparing them with normal BOEC, we were able to show

that, after endothelial cell activation by complement, platelet adhesion is at least

initially to VWF. It is known that platelets firmly adhere to endothelial cell-anchored

Page 96: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

84

VWF that has been unfurled under shear flow. This ensures platelets are brought in

close proximity to the injured endothelium, in order to initiate thrombosis and repair.

(De Ceunynck et al., 2013) Platelets also contain the complement regulator CFH and

it might be expected to aid in local complement control. (Licht et al., 2009)

There has always been considerable debate as to whether TTP and HUS are the same,

contiguous, or at two ends of a continuum or spectrum. Ultimately, with the discovery

of their respective causes, a dichotomy emerged, with one being considered as

complement-mediated and the other, VWF driven. (Remuzzi, 1987, 2003; Tsai, 2003)

Recently, however, those lines of separation were blurred again (Noris et al., 2012)

with the demonstration of complement activation in TTP patient‟s blood (Reti et al.,

2012; Ruiz-Torres et al., 2005; Wu et al., 2013) and the experimental association of

VWF with the alternative complement pathway. (Turner et al., 2014; Turner and

Moake, 2013)

Taken together this leads us to a new understanding of aHUS pathogenesis, or a two-

phase view of the sequence of events that might lead to TMA. Complement challenge

leads to endothelial cell activation with VWF release. This VWF facilitates platelet

adhesion and the binding of complement activation products. This brings platelets

into contact with the injured endothelium. In the presence of efficient and normal

levels of ADAMTS13 these VWF multimers, laden with complement products, are

cleaved and released into the circulation thus helping to maintain endothelial cell

homeostasis (Phase 1). If there is ongoing or persistent complement challenge,

especially in association with deficient or overpowered complement regulators, or less

VWF clearance (less blood flow or ADAMTS 13) then this initially protective effect

of VWF can be overcome, leading to progression towards a TMA phenotype (phase

2). (Figure 4-1) This might help explain the recent finding of ADAMTS13 mutations

in aHUS patients associated with decreased ADAMTS13 activity. (Feng et al., 2013a)

Page 97: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

85

Figure 4-1. VWF and ADAMTS13 as a Complement Regulatory System.

VWF released from EC upon complement activation is unfolded under shear flow allowing

the firm adhesion of platelets. VWF can also bind to complement activation products. In the

presence of normal ADAMTS13, then these multimers can be cleaved. VWF likely has a

complement regulatory role following its release from complement-activated EC.

Page 98: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

86

4.2 Future Directions

4.2.1 Introduction

After establishing BOEC as a model to study TMA pathogenesis we found that loss of

CD46/MCP function is not enough to fully explain the occurrence of TMA and that

additional complement aberrancies can significantly increase the likelihood of

progressing to a TMA phenotype. While seeking to better understand how a person

with a mutation in CD46/MCP may manifest a TMA, we next looked at VWF as a

potential amplifier of complement. This hypothesis arose from our own observation

of the association of immunofluorescently labeled VWF and C5b-9 under fluidic

conditions that coincided with a publication that demonstrated the association of

activated AP components on VWF. (Turner and Moake, 2013) Using a BOEC

naturally lacking VWF we actually found the contrary, that VWF more likely

downregulates complement. This exciting finding contradicts current published

opinion that VWF amplifies the complement injury (Turner et al., 2014) and opens up

a completely novel way of looking at the two diseases TTP and aHUS, perhaps

reopening the discussion as to how much they are linked.

Moving on from this, we aim to take a closer look at endothelial cell protection and

explaining how the cell protects itself from loss of CD46/MCP function as well as to

focus on more mechanistic details of complement-mediated EC injury. Specifically,

the model system can be employed to examine whether the cells mount a dynamic

response to complement-induced stress by increasing the expression of their surface

bound complement regulators. We have studied CD46/MCP as one of the two aHUS-

linked membrane-anchored complement regulators but we intentionally did not study

the contribution of the key soluble regulator CFH. To study its role is one of the key

next steps to complete the picture.

While addressing the example of CD46/MCP, we have utilized BOEC and thus

established a model system resembling patient characteristics in an unprecedented

way. Now we are in a position to test our findings in BOEC, not just from

CD46/MCP mutant patients, but to take it beyond that and extend our studies to study

BOEC from various subtypes of aHUS (and other TMA diseases) and to combine

them with patients own serum, plasma and platelets.

Page 99: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

87

4.2.2 Do Endothelial Cells Mount a Cytoprotective Response when

Challenged by Complement?

4.2.2.1 Rationale and Hypothesis

There appears to be a variation in the cell surface expression levels of CD46/MCP

and the other membrane-anchored complement regulators in various kidney diseases,

although the literature is inconsistent. In an in vitro study of cultured renal mesangial

cells exposed to either the anaphylatoxins C3a, C5a or the membrane attack complex

(MAC), a slight increase in surface expression of CD46/MCP was demonstrated by

flow cytometry although no change in mRNA could be demonstrated. In contrast, an

increase in CD55/DAF mRNA was observed. (Cosio et al., 1994) Another study of

human kidney tissue specimens found enhanced staining of CD46/MCP in association

with complement C3 split-product deposition in glomerulonephritic kidneys as

detected by immunohistochemistry. (Endoh et al., 1993)

Moreover, there is evidence to suggest that surface expression levels of complement

regulators can vary in vivo on different cell types and that this may play a role in the

etiology or susceptibility to disease including; (i) T-cell mediated rejection post

kidney transplant, (Kakuta et al., 2012) (ii) respiratory disorders,(Grumelli et al.,

2011; Lee et al., 2012) (iii) age-related macular degeneration,(Singh et al., 2012) (iv)

osteoarthritis, (Schulze-Tanzil et al., 2012; Scott et al., 2011) (v) rheumatoid

arthritis,(Pahwa et al., 2012) (vi) pregnancy related thrombophilia, (Wirstlein et al.,

2012) and (vii) systemic lupus erythematosus.(Alegretti et al., 2012; Das et al., 2013;

Ellinghaus et al., 2012) Expression levels of the regulators CD46/MCP and CD59

have been detected on neutrophils of patients after severe trauma. (Amara et al., 2010)

Furthermore in a model of STEC HUS, Shiga toxin 2 was shown to downregulate

expression of CD59 on the surface of glomerular endothelial and tubular epithelial

cells in vitro. (Ehrlenbach, Infect Immun 2013)

This suggests a hypothesis that when an EC is exposed to complement challenge, the

cellular response is to increase protector expression. If this dynamic response is

impaired by an endogenous defect or exogenous factor like toxins, then the cells are

more susceptible to injury.

Page 100: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

88

4.2.2.2 Methods

BOEC were seeded overnight in a 6 well plate (Falcon) to confluence, washed 1x

with HBSS before RNA was isolated using TRI Reagent (Sigma-Aldrich, St. Louis,

USA, T9424) according to manufacturer‟s instructions. RNA concentration and

integrity was verified by spectrophotometer (NanoDrop 1000, Thermo Fisher

Scientific, Waltham, USA), and reverse transcribed using ReadyScript™ cDNA

Synthesis Mix (Sigma-Aldrich, St. Louis, USA, RDRT). Samples (200 ng cDNA in

diethyl pyrocarbonate (DEPC, Sigma-Aldrich) treated water) were amplified by real

time polymerase chain reaction (PCR) using StepOne™ System from Life

Technologies (Carlsbad, USA). Amplified products were detected using KiCqStart™

SYBR® Green qPCR ReadyMix™, with ROX™ (Sigma-Aldrich, Carlsbad, USA,

KCQS02) and analyzed as follows: 2–(C

T – C

T GAPDH) – C

T control)

. GAPDH was used as

housekeeping gene.

The following oligonucleotide primers (Sigma-Aldrich, Carlsbad, USA) were used:

GAPDH: forward, 5‟-ACAGTTGCCATGTAGACC-3‟; reverse, 5‟-

TTTTTGGTTGAGCACAGG-3‟.

CD46/MCP: forward, 5‟-AGTGGTCAAATGTCGATTTC-3‟; reverse, 5‟-

ATCCCAAGTACTGTTACTGTC-3‟.

CD55/DAF: forward, 5‟-CAGAGGAAAATCTCTAACTTCC-3‟; reverse, 5‟-

AGTTGGTGAGACTTCTGTAG-3‟.

CD59: forward, 5‟-CATTACCAAAGCTGGGTTAC-3‟; reverse, 5‟-

TTTCTCTGATAAGGATGTCCC-3‟.

Using the following experimental conditions (each n = 3-4) where BOEC were

exposed to; (i) media (baseline), (ii) 50% NHS after CD46/MCP blockade and (iii)

50% NHS after CD46/MCP, CD55/DAF and CD59 blockade, for one, two and four

hours.

4.2.2.3 Preliminary Results

With CD46/MCP blockade alone, there was an initial increase in CD46/MCP

transcription, however, this did not persist, and no significant increase in either

Page 101: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

89

CD55/DAF, or CD59 was noted. (Figure 4-2 A) In contrast, by augmenting the

complement challenge with functional blockade of CD46/MCP, CD55/DAF and

CD59, BOEC increased their transcription of CD46/MCP and CD59. (Figure 4-2 B)

This was most prominent in the first 1-2 hours and the effect was lost at 4 hours.

Page 102: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

90

Figure 4-2. Response of complement regulators to increasing complement challenge

(A) BOEC were blocked with functional antibody against CD46/MCP for 30 minutes,

followed by incubation with 50% NHS in AP buffer for 1h, 2h and 4 hours. RNA was

isolated using TRI reagent and reverse transcribed to cDNA. CT values were normalized to

GAPDH expression and baseline (cells incubated with media). No significant difference in

gene expression of CD46, CD55 and CD59 up to 4 hours of treatment was observed, except

for an 18-fold increase of CD46 gene expression after one hour. N=3-4. (B) Gene expression

levels of CD46 and CD59 changed significantly within the first hour of complement

challenge when cells were blocked for CD46, CD55 and CD59. N=3-4. A two-way ANOVA

with Tukey‟s multiple comparison test was used for statistical analysis. * ≤0.05, ** ≤0.01,

***≤0.001, ****≤0.0001, ns > 0.05.

4.2.2.4 Discussion

In this preliminary study we see that with the increased complement challenge of

exposing BOEC to normal human serum after blocking three surface regulators as

compared to a single regulator, the mRNA expression of CD46/MCP and CD59

increase in the first one to two hours as evidence for dynamic response to protect the

EC. One study has looked at the expression levels of the other regulators CD55/DAF

and CD59 in eleven patients with CD46/MCP-associated aHUS and found no

difference in the expression levels in the patients as compared to healthy control

participants. The CD55/DAF and CD59 levels were measured on granulocytes,

however, and not endothelial cells. (Fremeaux-Bacchi et al., 2006)

Defining the response of normal EC to increasing complement challenge will be

important for when we study the BOEC of patients with disease, as an inability to

upregulate expression of the complement regulators might confer an increased risk of

manifesting disease.

Page 103: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

91

4.2.3 How Does the Fluid Phase Regulator CFH Contribute to Endothelial

Cell Protection?

4.2.3.1 Rationale and Hypothesis

All human cells are protected against complement-mediated injury of self by a

combination of fluid phase regulators, including complement Factor H (CFH) and

membrane-anchored regulators including CD46/Membrane Cofactor Protein (MCP),

CD55/Decay Accelerating Factor (DAF) and CD59. (Zipfel and Skerka, 2009) CFH

accelerates the decay of the AP C3 convertase and acts as a cofactor for the serine

protease CFI that cleaves C3b into inactive form iC3b. (Perkins et al., 2014) CFH is

considered the most important complement regulator. (Zipfel and Skerka, 2009) We

have utilized our model so far to demonstrate the effect of the membrane-anchored

complement regulators in cell protection. The effect of the other soluble regulators,

particularly CFH, will be important to analyze. Mutations in both CD46/MCP and

CFH have been described in individual patients presenting with aHUS. (Bresin et al.,

2013; Fremeaux-Bacchi et al., 2013)

The fact that CFH-related aHUS carries a worse prognosis than CD46/MCP with 75%

renal loss in the former as compared to 75% renal survival in the latter suggests that it

is the most important regulator for EC protection. (Fremeaux-Bacchi et al., 2013;

Noris et al., 2003; Noris et al., 2010; Sellier-Leclerc et al., 2007) Furthermore, an

additional CFH mutation in patients already carrying an MCP mutation increased the

likelihood of developing the disease and worsened the decline in renal function. We

postulate that there is a hierarchy of complement regulators and that loss of CFH on

top of CD46/MCP significantly increases the TMA phenotype.

4.2.3.2 Methods

CFH blocking

In order to study the effect of CFH in protecting the endothelium from cell death,

confluent BOEC were incubated with neuraminidase at a concentration of 30 mU/mL

in serum-free media for thirty minutes prior to 50% NHS. This disrupts the

glycocalyx and removes glycosaminoglycans (GAGs) by desialylation, as GAGs are

the physiological endothelial cells‟ binding site for CFH.(Junnikkala et al., 2000)

Page 104: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

92

Trypan blue exclusion

BOEC (between passages 3-14) were grown in 0.05 mg/ml rat-tail collagen type I

coated 96 well tissue culture plates (Sarstedt, Nümbrech, Germany). Cells were

incubated for thirty minutes at 37°C with (i) media, (ii) 50% normal human serum

(NHS) in AP buffer and (iii) 50% normal human serum (NHS) in AP buffer after pre-

incubation with functional blocking antibodies of CD46/MCP, CD55/DAF and CD59

in serum free EGM-2 media with supplements (Lonza). This treatment lasted thirty

minutes at 37°C. Then cells were washed twice with phosphate buffered saline (PBS:

Wisent, St Bruno, Canada). 1:1 mixture of Trypan Blue (Sigma-Aldrich, St. Louis,

USA) in PBS was added to cells for five minutes. 4% paraformaldehyde in PBS was

utilized to fix cells for ten minutes. Two fields of cells at 10x magnification from each

of duplicate wells were counted by using a Leitz DM IL microscope (Leica

Microsystems). The percent cell death was calculated by the following formula: dead

cells/total cells * 100.

Apoptosis

Cells were seeded in 96-well ELISA plates (Sarstedt) and incubated with functional

blocking antibodies for 30 minutes followed by 50% NHS in serum-free media

(cEGM-2) for four hours. Cells were fixed and blocked as described in detail earlier

and stained for cleaved caspase-3 (Cell Signaling, Danvers, USA, 1:400 dilution) and

respective secondary antibody conjugated with Alexa fluor 555 (1:1000 dilution).

4.2.3.3 Preliminary Results

We first examined whether neuraminidase (NA) pretreatment of cells followed by

exposure to normal human serum was associated with an increase in cell death over

serum alone. There was a three-fold higher increase in cell death with NA

pretreatment and 30 minutes of 50% NHS. (Figure 4-3 A) This cell death was shown

to be via apoptosis as assessed by fluorescently labeling fixed cells with an anti-

human cleaved caspase-3 antibody. There was an increase in the number of apoptotic

cells after 4 hours in those that had NA and an anti CD46 functional blockade prior to

serum.

Page 105: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

93

Figure 4-3. Role of CFH-mediated surface protection against endothelial cell death

(A) Neuraminidase (30mU) was used to mimic functional defect of CFH cell surface

protection. Cells grown to confluence on a 96-well plate were incubated for 30 minutes with

neuraminidase, followed by 30 minutes of 50% NHS/AP buffer. Trypan-blue was added and

positive cells were counted and displayed as percent of total cells. A significant (p=0.03, one-

way ANOVA) increase of dead cells was observed when compared to serum treatment alone.

N=5 (B-C) Apoptosis was determined using a cleaved-caspase 3 (1:400) antibody. Cells were

seeded in a 96-well plate, Neuraminidase (30mU) and a functional anti-CD46 antibody was

added for 30 minutes, followed by 4 hours of 50% NHS. Cells were fixed with 4% PFA and

stained for cleaved caspase 3 (red) and DAPI (blue). Cells were imaged using a Nikon Eclipse

Ti camera at 20x magnification (B) and 40x magnification (C).

Page 106: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

94

4.2.3.3 Discussion

These preliminary results confirm the importance of CFH as a regulator, and the

cumulative effect of a combined defect in CFH and CD46/MCP. However, because

thrombomodulin (CD141), also known to increase susceptibility to aHUS (Delvaeye

et al., 2009) is also a constituent of the endothelial cell glycocalyx, where it is bound

to chondroitin sulphate (Boels et al., 2013), neuraminidase might also be expected to

affect its function using this technique.(Boels et al., 2013) As such, this technique

may not be specific enough to clearly discriminate the precise CFH contribution.

Therefore, alternative approaches to more specifically assess the function of CFH

might include the use of CFH depleted serum or blockade of CFH function by either

(i) a CFH 19-20 construct, which binds and competitively inhibits the short consensus

repeats (SCR) 19-20 on the C-terminus of CFH, responsible for C3b binding

(Jokiranta et al., 2000; Jozsi et al., 2007) or (ii) a CFH-function blocking antibody.

Page 107: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

95

4.2.4 Will BOEC Isolated From a Patient Further our Understanding of

TMA?

4.2.4.1 Rationale, Hypothesis and Aims

Having established and validated the model system employing BOEC to study aHUS

and TMA, we are now poised to study BOEC isolated from patients with various

mutations and TMA diseases. The first patient that we will undertake this on carries

two heterozygous mutations in CD46/MCP and a heterozygous mutation in

ADAMTS13. (Figure 4-4) The affected proband manifested recurrent aHUS,

responsive to plasma therapy and eculizumab, when he inherited a heterozygous

mutation in CD46/MCP from his father and in CD46/MCP and ADAMTS13 from his

mother, in other words defects in both the complement and coagulation systems.

Family members carrying one or two mutations did not develop the disease so far.

Functional studies, carried out elsewhere, did not show a functional relevance for

either the CD46/MCP or the ADAMTS13 mutations alone and were not able to

explain disease manifestation in this patient. As the combination of inherited

alterations seem to have caused the disease in this individual, studying BOEC isolated

from the patient and relatives, might explain why the patient developed the disease

and his parents/siblings did not.

Page 108: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

96

Figure 4-4. Multiple genetic hits in complement and coagulation genes needed to cause

aHUS

The index patient (arrow) inherited two heterozygous mutations in MCP (blue, purple) and

one heterozygous mutation in ADAMTS13 (green). Other family members carry one or two

mutations, but never developed the disease.

4.2.4.3 Proposed Methods and Anticipated Results

BOEC that have been isolated from the patient will first be characterized as

endothelial, then as BOEC (positive by FACS for the endothelial markers CD144 and

CD31 and negative for the hemangioblast markers CD45 and CD14).

Immunofluorescence microscopic, FACS and qPCR examination of the expression of

the complement regulators as compared to normal control BOEC will then be

performed. The amount of complement deposition, cell survival and platelet adhesion

can then be compared to control healthy BOEC.

4.2.4.4 Anticipated Results and Implications

We anticipate that when these cells are exposed to normal human serum that there

may be increased complement deposition. Cell death and platelet adhesion assays will

possibly show an increased propensity to exhibit a TMA phenotype. Ultimately

isolating BOEC from patients will lead to a better understanding of the mechanisms

Page 109: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

97

involved in terms of disease susceptibility, manifestation or precipitation. It might

actually be able to explain why the patient developed the disease. Furthermore, in the

future isolating BOEC from patients might contribute to the pathogenetic workup of

aHUS patients. Finally, considering the regenerative capacity of BOEC and their use

as a means of cell therapy, they could become a treatment strategy.

Page 110: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

98

4.3 Conclusions

In 1924, Moschcowitz described a girl with microangiopathic hemolytic anemia,

thrombocytopenia, fever, neurological symptoms and hematuria. On post mortem

there were widespread, platelet-rich microthrombi in the microvasculature. This

became known as TTP. In 1955, Gasser, a Swiss pediatric nephrologist, described

patients with a similar presentation but in whom renal failure was prominent and he

termed this disease HUS. It also had a microangiopathic hemolytic anemia,

thrombocytopenia and microvascular thrombosis. Where the two diseases appeared to

differ, was whether the brain or the kidney was more prominently affected. (Gasser et

al., 1955) Over the next half century, as their respective etiologies became more clear,

they have been thought of, at various timepoints, and by various authors, as distinct,

separate and overlapping diseases. (Fujimura, 2003; Remuzzi, 1987, 2003; Tsai,

2003) In 1952, when Symmers coined the term thrombotic microangiopathy, it was

universally fatal. (Symmers, 1952) Although the mortality has significantly improved,

there remain many unmet needs in terms of understanding, specific therapies and

improving renal outcomes for individuals affected by this severe, multisystem disease.

In some ways, distinguishing TTP and aHUS is now more important than ever, as

there is a specific, complement-directed therapy, eculizumab that has revolutionized

the treatment for atypical HUS. (Legendre et al., 2013; Nurnberger et al., 2009)

As we continue to gain insight into aHUS, although a relatively rare disease, the

improved understanding will have significantly more broad-reaching implications for

a much larger group of patients, recognizing the increasing spectrum of diseases with

a TMA phenotype. (Riedl et al., 2014a) In the work presented in this thesis, we have

developed a system to study EC response to complement-mediated injury that can be

expanded upon and used to study the real EC phenotype of TMA patients as well as

model any of the diseases resulting in a TMA. We have confirmed that additional

genetic „hits‟ in complement significantly increase the likelihood of TMA over a

CD46/MCP mutation alone, and in so doing, have shown the importance of

biologically relevant model systems to study disease pathomechanisms. Most exciting

has been the discovery that VWF, released from activated ECs, is not complement

amplifying as proposed in the literature, but rather VWF and ADAMTS13 might well

be considered as down regulators of complement, where VWF laden with

complement can be cleaved by ADAMTS13 and released into the circulation for later

Page 111: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

99

clearance. In identifying VWF as complement regulator, this paves the way to a novel

understanding of aHUS pathogenesis. Firstly, complement challenged ECs release

VWF as a cell protective response - in order to bind platelets and help establish

control of complement activation. If this system is overcome, or if there is excessive

stimulation of the EC with loss of VWF, then the endothelium becomes more

vulnerable to complement injury. Thereafter platelets would be expected to adhere to

dead cells and exposed basement membrane.

Considering the heterogeneity of endothelial cell VWF expression and the relative

paucity of VWF in the glomerular endothelium, (Pusztaszeri et al., 2006) as opposed

to the brain microvascular endothelium, (Bernas et al., 2010; Dorovini-Zis et al.,

1991) this might explain why loss of complement control in aHUS particularly affects

the kidney microvasculature.

TMA pathogenesis involves the close interplay of a number of biological systems,

especially complement, coagulation and inflammation. At the center of this

interaction is the microvascular endothelium, and BOEC present a unique opportunity

to learn more about these rare and devastating diseases to ultimately develop safer,

more specific and efficient therapies. Finally, we believe that used as we have done,

BOEC will represent a unique toolset to perform functional studies of genes newly

discovered as linked to TMA.

Page 112: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

100

References

Ahmann, K.A., Johnson, S.L., Hebbel, R.P., and Tranquillo, R.T. (2011). Shear stress

responses of adult blood outgrowth endothelial cells seeded on bioartificial

tissue. Tissue Eng Part A 17, 2511-2521.

Aird, W.C. (2007a). Phenotypic heterogeneity of the endothelium: I. Structure,

function, and mechanisms. Circulation research 100, 158-173.

Aird, W.C. (2007b). Phenotypic heterogeneity of the endothelium: II.

Representative vascular beds. Circulation research 100, 174-190.

Alegretti, A.P., Schneider, L., Piccoli, A.K., Monticielo, O.A., Lora, P.S., Brenol, J.C.,

and Xavier, R.M. (2012). Diminished expression of complement regulatory

proteins on peripheral blood cells from systemic lupus erythematosus patients.

Clinical & developmental immunology 2012, 725684.

Amara, U., Kalbitz, M., Perl, M., Flierl, M.A., Rittirsch, D., Weiss, M., Schneider, M.,

Gebhard, F., and Huber-Lang, M. (2010). Early expression changes of

complement regulatory proteins and C5a receptor (CD88) on leukocytes after

multiple injury in humans. Shock 33, 568-575.

Amara, U., Rittirsch, D., Flierl, M., Bruckner, U., Klos, A., Gebhard, F., Lambris, J.D.,

and Huber-Lang, M. (2008). Interaction between the coagulation and

complement system. Advances in experimental medicine and biology 632, 71-79.

Barilla-LaBarca, M.L., Liszewski, M.K., Lambris, J.D., Hourcade, D., and Atkinson,

J.P. (2002). Role of membrane cofactor protein (CD46) in regulation of C4b and

C3b deposited on cells. J Immunol 168, 6298-6304.

Barre, P., Kaplan, B.S., de Chadarevian, J.P., and Drummond, K.N. (1977).

Hemolytic uremic syndrome with hypocomplementemia, serum C3NeF, and

glomerular deposits of C3. Archives of pathology & laboratory medicine 101,

357-361.

Bernas, M.J., Cardoso, F.L., Daley, S.K., Weinand, M.E., Campos, A.R., Ferreira, A.J.,

Hoying, J.B., Witte, M.H., Brites, D., Persidsky, Y., et al. (2010). Establishment of

primary cultures of human brain microvascular endothelial cells to provide an in

vitro cellular model of the blood-brain barrier. Nature protocols 5, 1265-1272.

Bienaime, F., Dragon-Durey, M.A., Regnier, C.H., Nilsson, S.C., Kwan, W.H., Blouin,

J., Jablonski, M., Renault, N., Rameix-Welti, M.A., Loirat, C., et al. (2010). Mutations

Page 113: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

101

in components of complement influence the outcome of Factor I-associated

atypical hemolytic uremic syndrome. Kidney international 77, 339-349.

Boels, M.G., Lee, D.H., van den Berg, B.M., Dane, M.J., van der Vlag, J., and Rabelink,

T.J. (2013). The endothelial glycocalyx as a potential modifier of the hemolytic

uremic syndrome. European journal of internal medicine 24, 503-509.

Bompais, H., Chagraoui, J., Canron, X., Crisan, M., Liu, X.H., Anjo, A., Tolla-Le Port,

C., Leboeuf, M., Charbord, P., Bikfalvi, A., et al. (2004). Human endothelial cells

derived from circulating progenitors display specific functional properties

compared with mature vessel wall endothelial cells. Blood 103, 2577-2584.

Brain, M.C. (1969). The haemolytic-uraemic syndrome. Seminars in hematology

6, 162-180.

Bresin, E., Rurali, E., Caprioli, J., Sanchez-Corral, P., Fremeaux-Bacchi, V.,

Rodriguez de Cordoba, S., Pinto, S., Goodship, T.H., Alberti, M., Ribes, D., et al.

(2013). Combined complement gene mutations in atypical hemolytic uremic

syndrome influence clinical phenotype. Journal of the American Society of

Nephrology : JASN 24, 475-486.

Brodbeck, W.G., Mold, C., Atkinson, J.P., and Medof, M.E. (2000). Cooperation

between decay-accelerating factor and membrane cofactor protein in protecting

cells from autologous complement attack. J Immunol 165, 3999-4006.

Bu, F., Maga, T., Meyer, N.C., Wang, K., Thomas, C.P., Nester, C.M., and Smith, R.J.

(2014). Comprehensive genetic analysis of complement and coagulation genes in

atypical hemolytic uremic syndrome. Journal of the American Society of

Nephrology : JASN 25, 55-64.

Camba, L., Al-Hilali, M.M., Shulji, M.M., Joyner, M.V., and Feest, T.G. (1985).

Haemolytic-uraemic syndrome with renal failure: the effect of plasmapheresis.

Haematologica 70, 341-344.

Campbell, W.D., Lazoura, E., Okada, N., and Okada, H. (2002). Inactivation of C3a

and C5a octapeptides by carboxypeptidase R and carboxypeptidase N.

Microbiology and immunology 46, 131-134.

Caprioli, J., Noris, M., Brioschi, S., Pianetti, G., Castelletti, F., Bettinaglio, P., Mele,

C., Bresin, E., Cassis, L., Gamba, S., et al. (2006). Genetics of HUS: the impact of

MCP, CFH, and IF mutations on clinical presentation, response to treatment, and

outcome. Blood 108, 1267-1279.

Page 114: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

102

Carreras, L., Romero, R., Requesens, C., Oliver, A.J., Carrera, M., Clavo, M., and

Alsina, J. (1981). Familial hypocomplementemic hemolytic uremic syndrome

with HLA-A3,B7 haplotype. JAMA : the journal of the American Medical

Association 245, 602-604.

Cataland, S.R., and Wu, H.M. (2014). Diagnosis and management of complement

mediated thrombotic microangiopathies. Blood reviews.

Chang Milbauer, L., Wei, P., Enenstein, J., Jiang, A., Hillery, C.A., Scott, J.P., Nelson,

S.C., Bodempudi, V., Topper, J.N., Yang, R.B., et al. (2008). Genetic endothelial

systems biology of sickle stroke risk. Blood 111, 3872-3879.

Clark, W.F. (2012). Thrombotic microangiopathy: current knowledge and

outcomes with plasma exchange. Semin Dial 25, 214-219.

Cole, J.L., Housley, G.A., Jr., Dykman, T.R., MacDermott, R.P., and Atkinson, J.P.

(1985). Identification of an additional class of C3-binding membrane proteins of

human peripheral blood leukocytes and cell lines. Proceedings of the National

Academy of Sciences of the United States of America 82, 859-863.

Cosio, F.G., Shibata, T., Rovin, B.H., and Birmingham, D.J. (1994). Effects of

complement activation products on the synthesis of decay accelerating factor

and membrane cofactor protein by human mesangial cells. Kidney international

46, 986-992.

Couzi, L., Contin-Bordes, C., Marliot, F., Sarrat, A., Grimal, P., Moreau, J.F., Merville,

P., and Fremeaux-Bacchi, V. (2008). Inherited deficiency of membrane cofactor

protein expression and varying manifestations of recurrent atypical hemolytic

uremic syndrome in a sibling pair. American journal of kidney diseases : the

official journal of the National Kidney Foundation 52, e5-9.

D'Angelo, A., Fattorini, A., and Crippa, L. (2009). Thrombotic microangiopathy in

pregnancy. Thrombosis research 123 Suppl 2, S56-62.

Das, N., Biswas, B., and Khera, R. (2013). Membrane-bound complement

regulatory proteins as biomarkers and potential therapeutic targets for SLE. In

Complement Therapeutics, J.D. Lambris, D. Ricklin, and V.M. Holers, eds. (233

Spring Street, New York NY 10013-1578, United States: Springer New York), pp.

55-81.

De Ceunynck, K., De Meyer, S.F., and Vanhoorelbeke, K. (2013). Unwinding the

von Willebrand factor strings puzzle. Blood 121, 270-277.

Page 115: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

103

De Ceunynck, K., Rocha, S., Feys, H.B., De Meyer, S.F., Uji-i, H., Deckmyn, H.,

Hofkens, J., and Vanhoorelbeke, K. (2011). Local elongation of endothelial cell-

anchored von Willebrand factor strings precedes ADAMTS13 protein-mediated

proteolysis. The Journal of biological chemistry 286, 36361-36367.

Deford, C.C., Reese, J.A., Schwartz, L.H., Perdue, J.J., Kremer Hovinga, J.A., Lammle,

B., Terrell, D.R., Vesely, S.K., and George, J.N. (2013). Multiple major morbidities

and increased mortality during long-term follow-up after recovery from

thrombotic thrombocytopenic purpura. Blood 122, 2023-2029; quiz 2142.

Degn, S.E., and Thiel, S. (2013). Humoral pattern recognition and the complement

system. Scandinavian journal of immunology 78, 181-193.

Delvaeye, M., and Conway, E.M. (2009). Coagulation and innate immune

responses: can we view them separately? Blood 114, 2367-2374.

Delvaeye, M., Noris, M., De Vriese, A., Esmon, C.T., Esmon, N.L., Ferrell, G., Del-

Favero, J., Plaisance, S., Claes, B., Lambrechts, D., et al. (2009). Thrombomodulin

mutations in atypical hemolytic-uremic syndrome. The New England journal of

medicine 361, 345-357.

Dong, J.F. (2005). Cleavage of ultra-large von Willebrand factor by ADAMTS-13

under flow conditions. J Thromb Haemost 3, 1710-1716.

Dong, J.F., Moake, J.L., Bernardo, A., Fujikawa, K., Ball, C., Nolasco, L., Lopez, J.A.,

and Cruz, M.A. (2003). ADAMTS-13 metalloprotease interacts with the

endothelial cell-derived ultra-large von Willebrand factor. The Journal of

biological chemistry 278, 29633-29639.

Dorovini-Zis, K., Prameya, R., and Bowman, P.D. (1991). Culture and

characterization of microvascular endothelial cells derived from human brain.

Laboratory investigation; a journal of technical methods and pathology 64, 425-

436.

Dragon-Durey, M.A., and Fremeaux-Bacchi, V. (2005). Atypical haemolytic

uraemic syndrome and mutations in complement regulator genes. Springer

seminars in immunopathology 27, 359-374.

Drukker, A., Winterborn, M., Bennett, B., Churg, J., Spitzer, A., and Greifer, I.

(1975). Recurrent hemolytic-uremic syndrome: a case report. Clinical

nephrology 4, 68-72.

Page 116: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

104

Ellinghaus, U., Pinder, C.L., Le Friec, G., Kemper, C., and Vyse, T.J. (2012). Severe

defects of complement regulator CD46-mediated IL-10 producing regulatory T

cells in patients with systemic lupus erythematosus. Immunology 137, 587.

Endoh, M., Yamashina, M., Ohi, H., Funahashi, K., Ikuno, T., Yasugi, T., Atkinson,

J.P., and Okada, H. (1993). Immunohistochemical demonstration of membrane

cofactor protein (MCP) of complement in normal and diseased kidney tissues.

Clinical and experimental immunology 94, 182-188.

Enenstein, J., Milbauer, L., Domingo, E., Wells, A., Roney, M., Kiley, J., Wei, P., and

Hebbel, R.P. (2010). Proinflammatory phenotype with imbalance of KLF2 and

RelA: risk of childhood stroke with sickle cell anemia. American journal of

hematology 85, 18-23.

Ensley, A.E., Nerem, R.M., Anderson, D.E., Hanson, S.R., and Hinds, M.T. (2012).

Fluid shear stress alters the hemostatic properties of endothelial outgrowth

cells. Tissue Eng Part A 18, 127-136.

Ermini, L., Goodship, T.H., Strain, L., Weale, M.E., Sacks, S.H., Cordell, H.J.,

Fremeaux-Bacchi, V., and Sheerin, N.S. (2012). Common genetic variants in

complement genes other than CFH, CD46 and the CFHRs are not associated with

aHUS. Molecular immunology 49, 640-648.

Esparza-Gordillo, J., Goicoechea de Jorge, E., Buil, A., Carreras Berges, L., Lopez-

Trascasa, M., Sanchez-Corral, P., and Rodriguez de Cordoba, S. (2005).

Predisposition to atypical hemolytic uremic syndrome involves the concurrence

of different susceptibility alleles in the regulators of complement activation gene

cluster in 1q32. Human molecular genetics 14, 703-712.

Esparza-Gordillo, J., Jorge, E.G., Garrido, C.A., Carreras, L., Lopez-Trascasa, M.,

Sanchez-Corral, P., and de Cordoba, S.R. (2006). Insights into hemolytic uremic

syndrome: segregation of three independent predisposition factors in a large,

multiple affected pedigree. Molecular immunology 43, 1769-1775.

Fearon, D.T., and Austen, K.F. (1975). Properdin: binding to C3b and stabilization

of the C3b-dependent C3 convertase. The Journal of experimental medicine 142,

856-863.

Feng, S., Eyler, S.J., Zhang, Y., Maga, T., Nester, C.M., Kroll, M.H., Smith, R.J., and

Afshar-Kharghan, V. (2013a). Partial ADAMTS13 deficiency in atypical hemolytic

uremic syndrome. Blood 122, 1487-1493.

Page 117: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

105

Feng, S., Kroll, M.H., Nolasco, L., Moake, J., and Afshar-Kharghan, V. (2013b).

Complement activation in thrombotic microangiopathies. British journal of

haematology 160, 404-406.

Feng, S., Liang, X., Cruz, M.A., Vu, H., Zhou, Z., Pemmaraju, N., Dong, J.F., Kroll,

M.H., and Afshar-Kharghan, V. (2013c). The interaction between factor H and

Von Willebrand factor. PloS one 8, e73715.

Fernandez, L.A., Sanz-Rodriguez, F., Zarrabeitia, R., Perez-Molino, A., Hebbel, R.P.,

Nguyen, J., Bernabeu, C., and Botella, L.M. (2005). Blood outgrowth endothelial

cells from Hereditary Haemorrhagic Telangiectasia patients reveal abnormalities

compatible with vascular lesions. Cardiovasc Res 68, 235-248.

Ferreira, V.P., Pangburn, M.K., and Cortes, C. (2010). Complement control protein

factor H: the good, the bad, and the inadequate. Molecular immunology 47, 2187-

2197.

Fremeaux-Bacchi, V., Arzouk, N., Ferlicot, S., Charpentier, B., Snanoudj, R., and

Durrbach, A. (2007). Recurrence of HUS due to CD46/MCP mutation after renal

transplantation: A role for endothelial microchimerism. American Journal of

Transplantation 7, 2047-2051.

Fremeaux-Bacchi, V., Fakhouri, F., Garnier, A., Bienaime, F., Dragon-Durey, M.A.,

Ngo, S., Moulin, B., Servais, A., Provot, F., Rostaing, L., et al. (2013). Genetics and

outcome of atypical hemolytic uremic syndrome: a nationwide French series

comparing children and adults. Clinical journal of the American Society of

Nephrology : CJASN 8, 554-562.

Fremeaux-Bacchi, V., Kemp, E.J., Goodship, J.A., Dragon-Durey, M.A., Strain, L.,

Loirat, C., Deng, H.W., and Goodship, T.H. (2005). The development of atypical

haemolytic-uraemic syndrome is influenced by susceptibility factors in factor H

and membrane cofactor protein: evidence from two independent cohorts.

Journal of medical genetics 42, 852-856.

Fremeaux-Bacchi, V., Moulton, E.A., Kavanagh, D., Dragon-Durey, M.A., Blouin, J.,

Caudy, A., Arzouk, N., Cleper, R., Francois, M., Guest, G., et al. (2006). Genetic and

functional analyses of membrane cofactor protein (CD46) mutations in atypical

hemolytic uremic syndrome. Journal of the American Society of Nephrology :

JASN 17, 2017-2025.

Page 118: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

106

Frimat, M., Tabarin, F., Dimitrov, J.D., Poitou, C., Halbwachs-Mecarelli, L.,

Fremeaux-Bacchi, V., and Roumenina, L.T. (2013). Complement activation by

heme as a secondary hit for atypical hemolytic uremic syndrome. Blood 122,

282-292.

Fuchs, S., Dohle, E., Kolbe, M., and Kirkpatrick, C.J. (2010). Outgrowth endothelial

cells: sources, characteristics and potential applications in tissue engineering

and regenerative medicine. Adv Biochem Eng Biotechnol 123, 201-217.

Fuchs, S., Hermanns, M.I., and Kirkpatrick, C.J. (2006). Retention of a

differentiated endothelial phenotype by outgrowth endothelial cells isolated

from human peripheral blood and expanded in long-term cultures. Cell Tissue

Res 326, 79-92.

Fujimura, Y. (2003). Is Upshaw-Schulman syndrome congenital thrombotic

thrombocytopenic purpura or hemolytic-uremic syndrome? Yes to both. J

Thromb Haemost 1, 2457-2458.

Furlan, M., Robles, R., Galbusera, M., Remuzzi, G., Kyrle, P.A., Brenner, B., Krause,

M., Scharrer, I., Aumann, V., Mittler, U., et al. (1998). von Willebrand factor-

cleaving protease in thrombotic thrombocytopenic purpura and the hemolytic-

uremic syndrome. The New England journal of medicine 339, 1578-1584.

Furlan, M., Robles, R., and Lammle, B. (1996). Partial purification and

characterization of a protease from human plasma cleaving von Willebrand

factor to fragments produced by in vivo proteolysis. Blood 87, 4223-4234.

Gasser, C., Gautier, E., Steck, A., Siebenmann, R.E., and Oechslin, R. (1955).

Hemolytic–uremic syndrome: bilateral necrosis of the renal cortex in acute

acquired hemolytic anemia. Schweizerische medizinische Wochenschrift, 905–

909.

Geerdink, L.M., Westra, D., van Wijk, J.A., Dorresteijn, E.M., Lilien, M.R., Davin, J.C.,

Komhoff, M., Van Hoeck, K., van der Vlugt, A., van den Heuvel, L.P., et al. (2012).

Atypical hemolytic uremic syndrome in children: complement mutations and

clinical characteristics. Pediatr Nephrol 27, 1283-1291.

George, J.N., Chen, Q., Deford, C.C., and Al-Nouri, Z. (2012). Ten patient stories

illustrating the extraordinarily diverse clinical features of patients with

thrombotic thrombocytopenic purpura and severe ADAMTS13 deficiency.

Journal of clinical apheresis 27, 302-311.

Page 119: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

107

George, J.N., and Nester, C.M. (2014). Syndromes of thrombotic microangiopathy.

The New England journal of medicine 371, 654-666.

Goodship, T.H. (2006). Atypical HUS and complement dysregulation. Journal of

the American Society of Nephrology : JASN 17, 1775-1776.

Goodship, T.H., Liszewski, M.K., Kemp, E.J., Richards, A., and Atkinson, J.P. (2004).

Mutations in CD46, a complement regulatory protein, predispose to atypical

HUS. Trends in molecular medicine 10, 226-231.

Gotze, O., and Muller-Eberhard, H.J. (1976). The alternative pathway of

complement activation. Advances in immunology 24, 1-35.

Gremmels, H., Fledderus, J.O., van Balkom, B.W., and Verhaar, M.C. (2011).

Transcriptome analysis in endothelial progenitor cell biology. Antioxidants &

redox signaling 15, 1029-1042.

Grumelli, S., Lu, B., Peterson, L., Maeno, T., and Gerard, C. (2011). CD46 protects

against chronic obstructive pulmonary disease. PloS one 6, e18785.

Hagge, W.W., Holley, K.E., Burke, E.C., and Stickler, G.B. (1967). Hemolytic-uremic

syndrome in two siblings. The New England journal of medicine 277, 138-139.

Harboe, M., and Mollnes, T.E. (2008). The alternative complement pathway

revisited. J Cell Mol Med 12, 1074-1084.

Harboe, M., Ulvund, G., Vien, L., Fung, M., and Mollnes, T.E. (2004). The

quantitative role of alternative pathway amplification in classical pathway

induced terminal complement activation. Clinical and experimental immunology

138, 439-446.

Hattori, R., Hamilton, K.K., McEver, R.P., and Sims, P.J. (1989). Complement

proteins C5b-9 induce secretion of high molecular weight multimers of

endothelial von Willebrand factor and translocation of granule membrane

protein GMP-140 to the cell surface. The Journal of biological chemistry 264,

9053-9060.

Hirschberg, R., Wang, S., and Mitu, G.M. (2008). Functional symbiosis between

endothelium and epithelial cells in glomeruli. Cell Tissue Res 331, 485-493.

Huber-Lang, M., Sarma, J.V., Zetoune, F.S., Rittirsch, D., Neff, T.A., McGuire, S.R.,

Lambris, J.D., Warner, R.L., Flierl, M.A., Hoesel, L.M., et al. (2006). Generation of

C5a in the absence of C3: a new complement activation pathway. Nature

medicine 12, 682-687.

Page 120: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

108

Ichida, S., Yuzawa, Y., Okada, H., Yoshioka, K., and Matsuo, S. (1994). Localization

of the complement regulatory proteins in the normal human kidney. Kidney

international 46, 89-96.

Jiang, A., Pan, W., Milbauer, L.C., Shyr, Y., and Hebbel, R.P. (2007). A practical

question based on cross-platform microarray data normalization: are BOEC

more like large vessel or microvascular endothelial cells or neither of them?

Journal of bioinformatics and computational biology 5, 875-893.

Jodele, S., Fukuda, T., Vinks, A., Mizuno, K., Laskin, B.L., Goebel, J., Dixon, B.P.,

Teusink, A., Pluthero, F.G., Lu, L., et al. (2014). Eculizumab therapy in children

with severe hematopoietic stem cell transplantation-associated thrombotic

microangiopathy. Biol Blood Marrow Transplant 20, 518-525.

Jodele, S., Licht, C., Goebel, J., Dixon, B.P., Zhang, K., Sivakumaran, T.A., Davies,

S.M., Pluthero, F.G., Lu, L., and Laskin, B.L. (2013). Abnormalities in the

alternative pathway of complement in children with hematopoietic stem cell

transplant-associated thrombotic microangiopathy. Blood 122, 2003-2007.

Johnson, S., and Waters, A. (2012). Is complement a culprit in infection-induced

forms of haemolytic uraemic syndrome? Immunobiology 217, 235-243.

Jokiranta, T.S., Hellwage, J., Koistinen, V., Zipfel, P.F., and Meri, S. (2000). Each of

the three binding sites on complement factor H interacts with a distinct site on

C3b. The Journal of biological chemistry 275, 27657-27662.

Jozsi, M., Oppermann, M., Lambris, J.D., and Zipfel, P.F. (2007). The C-terminus of

complement factor H is essential for host cell protection. Molecular immunology

44, 2697-2706.

Junnikkala, S., Jokiranta, T.S., Friese, M.A., Jarva, H., Zipfel, P.F., and Meri, S.

(2000). Exceptional resistance of human H2 glioblastoma cells to complement-

mediated killing by expression and utilization of factor H and factor H-like

protein 1. J Immunol 164, 6075-6081.

Kahlenberg, J.M., Thacker, S.G., Berthier, C.C., Cohen, C.D., Kretzler, M., and

Kaplan, M.J. (2011). Inflammasome activation of IL-18 results in endothelial

progenitor cell dysfunction in systemic lupus erythematosus. J Immunol 187,

6143-6156.

Kakuta, Y., Okumi, M., Kato, T., Tsutahara, K., Yazawa, K., Takahara, S., and

Nonomura, N. (2012). Expression of the membrane cofactor protein in renal

Page 121: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

109

allografts during acute T-cell-mediated rejection. Journal of Urology 1), e914-

e915.

Kaneko, Y., Tajiri, N., Shinozuka, K., Glover, L.E., Weinbren, N.L., Cortes, L., and

Borlongan, C.V. (2012). Cell therapy for stroke: emphasis on optimizing safety

and efficacy profile of endothelial progenitor cells. Current pharmaceutical

design 18, 3731-3734.

Kaplan, B.S. (1977). Hemolytic uremic syndrome with recurrent episodes: an

important subset. Clinical nephrology 8, 495-498.

Kaplan, B.S., Chesney, R.W., and Drummond, K.N. (1975). Hemolytic uremic

syndrome in families. The New England journal of medicine 292, 1090-1093.

Karmali, M.A., Steele, B.T., Petric, M., and Lim, C. (1983). Sporadic cases of

haemolytic-uraemic syndrome associated with faecal cytotoxin and cytotoxin-

producing Escherichia coli in stools. Lancet 1, 619-620.

Kavanagh, D., Goodship, T.H., and Richards, A. (2006). Atypical haemolytic

uraemic syndrome. British medical bulletin 77-78, 5-22.

Kavanagh, D., Kemp, E.J., Mayland, E., Winney, R.J., Duffield, J.S., Warwick, G.,

Richards, A., Ward, R., Goodship, J.A., and Goodship, T.H. (2005). Mutations in

complement factor I predispose to development of atypical hemolytic uremic

syndrome. Journal of the American Society of Nephrology : JASN 16, 2150-2155.

Kawanami, O., Jin, E., Ghazizadeh, M., Fujiwara, M., Jiang, L., Nagashima, M.,

Shimizu, H., Takemura, T., Ohaki, Y., Arai, S., et al. (2000). Heterogeneous

distribution of thrombomodulin and von Willebrand factor in endothelial cells in

the human pulmonary microvessels. J Nippon Med Sch 67, 118-125.

Kinoshita, S., Yoshioka, A., Park, Y.D., Ishizashi, H., Konno, M., Funato, M., Matsui,

T., Titani, K., Yagi, H., Matsumoto, M., et al. (2001). Upshaw-Schulman syndrome

revisited: a concept of congenital thrombotic thrombocytopenic purpura.

International journal of hematology 74, 101-108.

Knobl, P. (2014). Inherited and acquired thrombotic thrombocytopenic purpura

(TTP) in adults. Seminars in thrombosis and hemostasis 40, 493-502.

Kojouri, K., Vesely, S.K., and George, J.N. (2001). Quinine-associated thrombotic

thrombocytopenic purpura-hemolytic uremic syndrome: frequency, clinical

features, and long-term outcomes. Annals of internal medicine 135, 1047-1051.

Page 122: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

110

Krisinger, M.J., Goebeler, V., Lu, Z., Meixner, S.C., Myles, T., Pryzdial, E.L., and

Conway, E.M. (2012). Thrombin generates previously unidentified C5 products

that support the terminal complement activation pathway. Blood 120, 1717-

1725.

Kwaan, H.C. (2011). Microvascular thrombosis: a serious and deadly pathologic

process in multiple diseases. Seminars in thrombosis and hemostasis 37, 961-

978.

Lancellotti, S., and De Cristofaro, R. (2011). Structure and proteolytic properties

of ADAMTS13, a metalloprotease involved in the pathogenesis of thrombotic

microangiopathies. Prog Mol Biol Transl Sci 99, 105-144.

Lee, J.S., Bae, J.S., Kim, J.H., Kim, J.Y., Park, T.J., Pasaje, C.F., Park, B.L., Cheong, H.S.,

Uh, S.T., Jang, A.S., et al. (2012). CD55 polymorphisms and risk of aspirin-

exacerbated respiratory disease. Molecular Medicine Reports 6, 1087-1092.

Legendre, C.M., Licht, C., Muus, P., Greenbaum, L.A., Babu, S., Bedrosian, C.,

Bingham, C., Cohen, D.J., Delmas, Y., Douglas, K., et al. (2013). Terminal

complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. The

New England journal of medicine 368, 2169-2181.

Lemaire, M., Fremeaux-Bacchi, V., Schaefer, F., Choi, M., Tang, W.H., Le Quintrec,

M., Fakhouri, F., Taque, S., Nobili, F., Martinez, F., et al. (2013). Recessive

mutations in DGKE cause atypical hemolytic-uremic syndrome. Nature genetics

45, 531-536.

Levy, G.G., Nichols, W.C., Lian, E.C., Foroud, T., McClintick, J.N., McGee, B.M., Yang,

A.Y., Siemieniak, D.R., Stark, K.R., Gruppo, R., et al. (2001). Mutations in a member

of the ADAMTS gene family cause thrombotic thrombocytopenic purpura. Nature

413, 488-494.

Licht, C., Pluthero, F.G., Li, L., Christensen, H., Habbig, S., Hoppe, B., Geary, D.F.,

Zipfel, P.F., and Kahr, W.H. (2009). Platelet-associated complement factor H in

healthy persons and patients with atypical HUS. Blood 114, 4538-4545.

Lin, Y., Weisdorf, D.J., Solovey, A., and Hebbel, R.P. (2000). Origins of circulating

endothelial cells and endothelial outgrowth from blood. The Journal of clinical

investigation 105, 71-77.

Page 123: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

111

Liszewski, M.K., and Atkinson, J.P. (1996). Membrane cofactor protein (MCP;

CD46). Isoforms differ in protection against the classical pathway of

complement. J Immunol 156, 4415-4421.

Liszewski, M.K., Fang, C.J., and Atkinson, J.P. (2008). Inhibiting complement

activation on cells at the step of C3 cleavage. Vaccine 26 Suppl 8, I22-27.

Liszewski, M.K., Leung, M., Cui, W., Subramanian, V.B., Parkinson, J., Barlow, P.N.,

Manchester, M., and Atkinson, J.P. (2000). Dissecting sites important for

complement regulatory activity in membrane cofactor protein (MCP; CD46). The

Journal of biological chemistry 275, 37692-37701.

Liszewski, M.K., Leung, M.K., Schraml, B., Goodship, T.H., and Atkinson, J.P.

(2007). Modeling how CD46 deficiency predisposes to atypical hemolytic uremic

syndrome. Molecular immunology 44, 1559-1568.

Liszewski, M.K., Post, T.W., and Atkinson, J.P. (1991). Membrane cofactor protein

(MCP or CD46): newest member of the regulators of complement activation gene

cluster. Annual review of immunology 9, 431-455.

Lopez, J.A., and Dong, J.F. (2004). Cleavage of von Willebrand factor by ADAMTS-

13 on endothelial cells. Seminars in hematology 41, 15-23.

Louise, C.B., and Obrig, T.G. (1994). Human renal microvascular endothelial cells

as a potential target in the development of the hemolytic uremic syndrome as

related to fibrinolysis factor expression, in vitro. Microvasc Res 47, 377-387.

Maga, T.K., Nishimura, C.J., Weaver, A.E., Frees, K.L., and Smith, R.J. (2010).

Mutations in alternative pathway complement proteins in American patients

with atypical hemolytic uremic syndrome. Human mutation 31, E1445-1460.

Maisner, A., Liszewski, M.K., Atkinson, J.P., Schwartz-Albiez, R., and Herrler, G.

(1996). Two different cytoplasmic tails direct isoforms of the membrane cofactor

protein (CD46) to the basolateral surface of Madin-Darby canine kidney cells.

The Journal of biological chemistry 271, 18853-18858.

Malina, M., Roumenina, L.T., Seeman, T., Le Quintrec, M., Dragon-Durey, M.A.,

Schaefer, F., and Fremeaux-Bacchi, V. (2012). Genetics of hemolytic uremic

syndromes. Presse Med 41, e105-114.

Marinozzi, M.C., Vergoz, L., Rybkine, T., Ngo, S., Bettoni, S., Pashov, A., Cayla, M.,

Tabarin, F., Jablonski, M., Hue, C., et al. (2014). Complement Factor B Mutations

Page 124: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

112

in Atypical Hemolytic Uremic Syndrome--Disease-Relevant or Benign? Journal of

the American Society of Nephrology : JASN.

Markiewski, M.M., Nilsson, B., Nilsson Ekdahl, K., Mollnes, T.E., and Lambris, J.D.

(2007). Complement and coagulation: strangers or partners in crime? Trends in

immunology 28, 184-192.

Martin-Ramirez, J., Hofman, M., van den Biggelaar, M., Hebbel, R.P., and Voorberg,

J. (2012). Establishment of outgrowth endothelial cells from peripheral blood.

Nature protocols 7, 1709-1715.

Martinez-Barricarte, R., Pianetti, G., Gautard, R., Misselwitz, J., Strain, L.,

Fremeaux-Bacchi, V., Skerka, C., Zipfel, P.F., Goodship, T., Noris, M., et al. (2008).

The complement factor H R1210C mutation is associated with atypical hemolytic

uremic syndrome. Journal of the American Society of Nephrology : JASN 19, 639-

646.

Matsushita, M., Endo, Y., and Fujita, T. (2013). Structural and functional overview

of the lectin complement pathway: its molecular basis and physiological

implication. Archivum immunologiae et therapiae experimentalis 61, 273-283.

Mazzolai, L., Bouzourene, K., Hayoz, D., Dignat-George, F., Liu, J.W., Bounameaux,

H., Dunoyer-Geindre, S., and Kruithof, E.K. (2011). Characterization of human late

outgrowth endothelial progenitor-derived cells under various flow conditions. J

Vasc Res 48, 443-451.

McNearney, T., Ballard, L., Seya, T., and Atkinson, J.P. (1989). Membrane cofactor

protein of complement is present on human fibroblast, epithelial, and endothelial

cells. The Journal of clinical investigation 84, 538-545.

Medina, R.J., O'Neill, C.L., Humphreys, M.W., Gardiner, T.A., and Stitt, A.W.

(2010a). Outgrowth endothelial cells: characterization and their potential for

reversing ischemic retinopathy. Invest Ophthalmol Vis Sci 51, 5906-5913.

Medina, R.J., O'Neill, C.L., O'Doherty, T.M., Wilson, S.E., and Stitt, A.W. (2012).

Endothelial progenitors as tools to study vascular disease. Stem Cells Int 2012,

346735.

Medina, R.J., O'Neill, C.L., Sweeney, M., Guduric-Fuchs, J., Gardiner, T.A., Simpson,

D.A., and Stitt, A.W. (2010b). Molecular analysis of endothelial progenitor cell

(EPC) subtypes reveals two distinct cell populations with different identities.

BMC Med Genomics 3, 18.

Page 125: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

113

Miller, B.W., Hmiel, S.P., Schnitzler, M.A., and Brennan, D.C. (1997). Cyclosporine

as cause of thrombotic microangiopathy after renal transplantation. American

journal of kidney diseases : the official journal of the National Kidney Foundation

29, 813-814.

Moake, J.L., Rudy, C.K., Troll, J.H., Weinstein, M.J., Colannino, N.M., Azocar, J.,

Seder, R.H., Hong, S.L., and Deykin, D. (1982). Unusually large plasma factor

VIII:von Willebrand factor multimers in chronic relapsing thrombotic

thrombocytopenic purpura. The New England journal of medicine 307, 1432-

1435.

Moschcowitz, E. (1924). Hyaline thrombosis of the terminal arterioles and

capillaries: a hitherto undescribed disease. Proc NY Pathol Soc, 21-24.

Muller-Eberhard, H.J. (1986). The membrane attack complex of complement.

Annual review of immunology 4, 503-528.

Muller-Eberhard, H.J. (1988). Molecular organization and function of the

complement system. Annu Rev Biochem 57, 321-347.

Muller, A.M., Skrzynski, C., Skipka, G., and Muller, K.M. (2002). Expression of von

Willebrand factor by human pulmonary endothelial cells in vivo. Respiration 69,

526-533.

Nakanishi, I., Moutabarrik, A., Hara, T., Hatanaka, M., Hayashi, T., Syouji, T.,

Okada, N., Kitamura, E., Tsubakihara, Y., Matsumoto, M., et al. (1994).

Identification and characterization of membrane cofactor protein (CD46) in the

human kidneys. European journal of immunology 24, 1529-1535.

Nakashima, S., Qian, Z., Rahimi, S., Wasowska, B.A., and Baldwin, W.M., 3rd

(2002). Membrane attack complex contributes to destruction of vascular

integrity in acute lung allograft rejection. J Immunol 169, 4620-4627.

Nightingale, T., and Cutler, D. (2013). The secretion of von Willebrand factor

from endothelial cells; an increasingly complicated story. J Thromb Haemost 11

Suppl 1, 192-201.

Nolasco, L., Nolasco, J., Feng, S., Afshar-Kharghan, V., and Moake, J. (2013).

Human complement factor H is a reductase for large soluble von Willebrand

factor multimers--brief report. Arterioscler Thromb Vasc Biol 33, 2524-2528.

Nonaka, M. (2014). Evolution of the complement system. Sub-cellular

biochemistry 80, 31-43.

Page 126: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

114

Noone, D., Al-Matrafi, J., Tinckam, K., Zipfel, P.F., Herzenberg, A.M., Thorner, P.S.,

Pluthero, F.G., Kahr, W.H., Filler, G., Hebert, D., et al. (2012). Antibody mediated

rejection associated with complement factor h-related protein 3/1 deficiency

successfully treated with eculizumab. American journal of transplantation :

official journal of the American Society of Transplantation and the American

Society of Transplant Surgeons 12, 2546-2553.

Noris, M., Brioschi, S., Caprioli, J., Todeschini, M., Bresin, E., Porrati, F., Gamba, S.,

and Remuzzi, G. (2003). Familial haemolytic uraemic syndrome and an MCP

mutation. Lancet 362, 1542-1547.

Noris, M., Caprioli, J., Bresin, E., Mossali, C., Pianetti, G., Gamba, S., Daina, E., Fenili,

C., Castelletti, F., Sorosina, A., et al. (2010). Relative role of genetic complement

abnormalities in sporadic and familial aHUS and their impact on clinical

phenotype. Clinical journal of the American Society of Nephrology : CJASN 5,

1844-1859.

Noris, M., Mescia, F., and Remuzzi, G. (2012). STEC-HUS, atypical HUS and TTP

are all diseases of complement activation. Nature reviews Nephrology 8, 622-

633.

Noris, M., and Remuzzi, G. (2009). Atypical hemolytic-uremic syndrome. The

New England journal of medicine 361, 1676-1687.

Noris, M., Ruggenenti, P., Perna, A., Orisio, S., Caprioli, J., Skerka, C., Vasile, B.,

Zipfel, P.F., and Remuzzi, G. (1999). Hypocomplementemia discloses genetic

predisposition to hemolytic uremic syndrome and thrombotic thrombocytopenic

purpura: role of factor H abnormalities. Italian Registry of Familial and

Recurrent Hemolytic Uremic Syndrome/Thrombotic Thrombocytopenic

Purpura. Journal of the American Society of Nephrology : JASN 10, 281-293.

Nurnberger, J., Philipp, T., Witzke, O., Opazo Saez, A., Vester, U., Baba, H.A.,

Kribben, A., Zimmerhackl, L.B., Janecke, A.R., Nagel, M., et al. (2009). Eculizumab

for atypical hemolytic-uremic syndrome. The New England journal of medicine

360, 542-544.

Oikonomopoulou, K., Ricklin, D., Ward, P.A., and Lambris, J.D. (2012).

Interactions between coagulation and complement--their role in inflammation.

Seminars in immunopathology 34, 151-165.

Page 127: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

115

Ota, H., Fox-Talbot, K., Hu, W., Qian, Z., Sanfilippo, F., Hruban, R.H., and Baldwin,

W.M., 3rd (2005). Terminal complement components mediate release of von

Willebrand factor and adhesion of platelets in arteries of allografts.

Transplantation 79, 276-281.

Othman, M., Chirinian, Y., Brown, C., Notley, C., Hickson, N., Hampshire, D.,

Buckley, S., Waddington, S., Parker, A.L., Baker, A., et al. (2010). Functional

characterization of a 13-bp deletion (c.-1522_-1510del13) in the promoter of the

von Willebrand factor gene in type 1 von Willebrand disease. Blood 116, 3645-

3652.

Pahwa, R., Kumar, U., and Das, N. (2012). Role of complement regulatory protein

CD46 in rheumatoid arthritis. Immunology 137, 197.

Pangburn, M.K., Ferreira, V.P., and Cortes, C. (2008). Discrimination between host

and pathogens by the complement system. Vaccine 26 Suppl 8, I15-21.

Perkins, S.J., Fung, K.W., and Khan, S. (2014). Molecular Interactions between

Complement Factor H and Its Heparin and Heparan Sulfate Ligands. Frontiers in

immunology 5, 126.

Pichette, V., Querin, S., Schurch, W., Brun, G., Lehner-Netsch, G., and Delage, J.M.

(1994). Familial hemolytic-uremic syndrome and homozygous factor H

deficiency. American journal of kidney diseases : the official journal of the

National Kidney Foundation 24, 936-941.

Pillemer, L., Blum, L., Lepow, I.H., Ross, O.A., Todd, E.W., and Wardlaw, A.C.

(1954). The properdin system and immunity. I. Demonstration and isolation of a

new serum protein, properdin, and its role in immune phenomena. Science 120,

279-285.

Pirson, Y., Lefebvre, C., Arnout, C., and van Ypersele de Strihou, C. (1987).

Hemolytic uremic syndrome in three adult siblings: a familial study and

evolution. Clinical nephrology 28, 250-255.

Post, T.W., Liszewski, M.K., Adams, E.M., Tedja, I., Miller, E.A., and Atkinson, J.P.

(1991). Membrane cofactor protein of the complement system: alternative

splicing of serine/threonine/proline-rich exons and cytoplasmic tails produces

multiple isoforms that correlate with protein phenotype. The Journal of

experimental medicine 174, 93-102.

Page 128: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

116

Provaznikova, D., Rittich, S., Malina, M., Seeman, T., Marinov, I., Riedl, M., and

Hrachovinova, I. (2012). Manifestation of atypical hemolytic uremic syndrome

caused by novel mutations in MCP. Pediatric Nephrology 27, 73-81.

Pusztaszeri, M.P., Seelentag, W., and Bosman, F.T. (2006). Immunohistochemical

expression of endothelial markers CD31, CD34, von Willebrand factor, and Fli-1

in normal human tissues. The journal of histochemistry and cytochemistry :

official journal of the Histochemistry Society 54, 385-395.

Radhi, M., and Carpenter, S.L. (2012). Thrombotic microangiopathies. ISRN

Hematol 2012, 310596.

Ray, P.E., Tassi, E., Liu, X.H., and Wellstein, A. (2006). Role of fibroblast growth

factor-binding protein in the pathogenesis of HIV-associated hemolytic uremic

syndrome. Am J Physiol Regul Integr Comp Physiol 290, R105-113.

Rayes, J., Roumenina, L.T., Dimitrov, J.D., Repesse, Y., Ing, M., Christophe, O.,

Jokiranta, T.S., Halbwachs-Mecarelli, L., Borel-Derlon, A., Kaveri, S.V., et al.

(2014). The interaction between factor H and VWF increases factor H cofactor

activity and regulates VWF prothrombotic status. Blood 123, 121-125.

Reinisch, A., Hofmann, N.A., Obenauf, A.C., Kashofer, K., Rohde, E., Schallmoser, K.,

Flicker, K., Lanzer, G., Linkesch, W., Speicher, M.R., et al. (2009). Humanized

large-scale expanded endothelial colony-forming cells function in vitro and in

vivo. Blood 113, 6716-6725.

Remuzzi, G. (1987). HUS and TTP: variable expression of a single entity. Kidney

international 32, 292-308.

Remuzzi, G. (2003). Is ADAMTS-13 deficiency specific for thrombotic

thrombocytopenic purpura? No. J Thromb Haemost 1, 632-634.

Remuzzi, G., Misiani, R., Marchesi, D., Livio, M., Mecca, G., de Gaetano, G., and

Donati, M.B. (1978). Haemolytic-uraemic syndrome: deficiency of plasma

factor(s) regulating prostacyclin activity? Lancet 2, 871-872.

Remuzzi, G., Misiani, R., Marchesi, D., Livio, M., Mecca, G., de Gaetano, G., and

Donati, M.D. (1979). Treatment of the hemolytic uremic syndrome with plasma.

Clinical nephrology 12, 279-284.

Remuzzi, G., and Ruggenenti, P. (1992). Plasma manipulation in hemolytic

uremic syndrome and thrombotic thrombocytopenic purpura. Annales de

medecine interne 143 Suppl 1, 19-26.

Page 129: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

117

Rennard, S., and Abe, S. (1979). Decreased cold-insoluble globulin in congenital

thrombocytopenia (Upshaw-Schulman syndrome). The New England journal of

medicine 300, 368.

Reti, M., Farkas, P., Csuka, D., Razso, K., Schlammadinger, A., Udvardy, M.L.,

Madach, K., Domjan, G., Bereczki, C., Reusz, G.S., et al. (2012). Complement

activation in thrombotic thrombocytopenic purpura. J Thromb Haemost 10, 791-

798.

Richards, A., Kathryn Liszewski, M., Kavanagh, D., Fang, C.J., Moulton, E.,

Fremeaux-Bacchi, V., Remuzzi, G., Noris, M., Goodship, T.H.J., and Atkinson, J.P.

(2007). Implications of the initial mutations in membrane cofactor protein (MCP;

CD46) leading to atypical hemolytic uremic syndrome. Molecular immunology

44, 111-122.

Richards, A., Kemp, E.J., Liszewski, M.K., Goodship, J.A., Lampe, A.K., Decorte, R.,

Muslumanoglu, M.H., Kavukcu, S., Filler, G., Pirson, Y., et al. (2003). Mutations in

human complement regulator, membrane cofactor protein (CD46), predispose to

development of familial hemolytic uremic syndrome. Proceedings of the National

Academy of Sciences of the United States of America 100, 12966-12971.

Ricklin, D., Hajishengallis, G., Yang, K., and Lambris, J.D. (2010). Complement: a

key system for immune surveillance and homeostasis. Nature immunology 11,

785-797.

Riedl, M., Fakhouri, F., Le Quintrec, M., Noone, D.G., Jungraithmayr, T.C.,

Fremeaux-Bacchi, V., and Licht, C. (2014a). Spectrum of complement-mediated

thrombotic microangiopathies: pathogenetic insights identifying novel treatment

approaches. Seminars in thrombosis and hemostasis 40, 444-464.

Riedl, M., Orth-Holler, D., and Wurzner, R. (2014b). An update on the thrombotic

microangiopathies hemolytic uremic syndrome (HUS) and thrombotic

thrombocytopenic purpura (TTP). Seminars in thrombosis and hemostasis 40,

413-415.

Rignault-Clerc, S., Bielmann, C., Delodder, F., Raffoul, W., Waeber, B., Liaudet, L.,

Berger, M.M., Feihl, F., and Rosenblatt-Velin, N. (2012). Functional late outgrowth

endothelial progenitors isolated from peripheral blood of burned patients. Burns

: journal of the International Society for Burn Injuries.

Page 130: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

118

Rodriguez de Cordoba, S., Diaz-Guillen, M.A., and Heine-Suner, D. (1999). An

integrated map of the human regulator of complement activation (RCA) gene

cluster on 1q32. Molecular immunology 36, 803-808.

Ruggeri, Z.M. (2007). The role of von Willebrand factor in thrombus formation.

Thrombosis research 120 Suppl 1, S5-9.

Ruiz-Torres, M.P., Casiraghi, F., Galbusera, M., Macconi, D., Gastoldi, S.,

Todeschini, M., Porrati, F., Belotti, D., Pogliani, E.M., Noris, M., et al. (2005).

Complement activation: the missing link between ADAMTS-13 deficiency and

microvascular thrombosis of thrombotic microangiopathies. Thrombosis and

haemostasis 93, 443-452.

Sasahara, Y., Kumaki, S., Ohashi, Y., Minegishi, M., Kano, H., Bessho, F., and

Tsuchiya, S. (2001). Deficient activity of von Willebrand factor-cleaving protease

in patients with Upshaw-Schulman syndrome. International journal of

hematology 74, 109-114.

Satoskar, A.A., Pelletier, R., Adams, P., Nadasdy, G.M., Brodsky, S., Pesavento, T.,

Henry, M., and Nadasdy, T. (2010). De novo thrombotic microangiopathy in renal

allograft biopsies-role of antibody-mediated rejection. American journal of

transplantation : official journal of the American Society of Transplantation and

the American Society of Transplant Surgeons 10, 1804-1811.

Scheiring, J., Andreoli, S.P., and Zimmerhackl, L.B. (2008). Treatment and

outcome of Shiga-toxin-associated hemolytic uremic syndrome (HUS). Pediatr

Nephrol 23, 1749-1760.

Schiff, D.E., Roberts, W.D., Willert, J., and Tsai, H.M. (2004). Thrombocytopenia

and severe hyperbilirubinemia in the neonatal period secondary to congenital

thrombotic thrombocytopenic purpura and ADAMTS13 deficiency. Journal of

pediatric hematology/oncology 26, 535-538.

Schneider, S.W., Nuschele, S., Wixforth, A., Gorzelanny, C., Alexander-Katz, A.,

Netz, R.R., and Schneider, M.F. (2007). Shear-induced unfolding triggers adhesion

of von Willebrand factor fibers. Proceedings of the National Academy of Sciences

of the United States of America 104, 7899-7903.

Schulze-Tanzil, G., Kohl, B., Sayed, K.E., Arens, S., Ertel, W., Stolzel, K., and John, T.

(2012). Anaphylatoxin receptors and complement regulatory proteins in human

Page 131: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

119

articular and non-articular chondrocytes: Interrelation with cytokines. Cell and

Tissue Research 350, 465-475.

Scott, J.L., Scanzello, C.R., and Crow, M.K. (2011). Membrane-bound complement

regulatory protein expression decreases in synovium during OA progression and

is associated with increased C5b-9 deposition. Osteoarthritis and Cartilage 19,

S55.

Sellier-Leclerc, A.L., Fremeaux-Bacchi, V., Dragon-Durey, M.A., Macher, M.A.,

Niaudet, P., Guest, G., Boudailliez, B., Bouissou, F., Deschenes, G., Gie, S., et al.

(2007). Differential impact of complement mutations on clinical characteristics

in atypical hemolytic uremic syndrome. Journal of the American Society of

Nephrology : JASN 18, 2392-2400.

Seya, T., and Atkinson, J.P. (1989). Functional properties of membrane cofactor

protein of complement. The Biochemical journal 264, 581-588.

Seya, T., Ballard, L.L., Bora, N.S., Kumar, V., Cui, W., and Atkinson, J.P. (1988).

Distribution of membrane cofactor protein of complement on human peripheral

blood cells. An altered form is found on granulocytes. European journal of

immunology 18, 1289-1294.

Seya, T., Turner, J.R., and Atkinson, J.P. (1986). Purification and characterization

of a membrane protein (gp45-70) that is a cofactor for cleavage of C3b and C4b.

The Journal of experimental medicine 163, 837-855.

Singh, A., Faber, C., Falk, M., Nissen, M.H., Hviid, T.V.F., and Sorensen, T.L. (2012).

Altered expression of CD46 and CD59 on leukocytes in neovascular age-related

macular degeneration. American Journal of Ophthalmology 154, 193-199.

Skidgel, R.A., and Erdos, E.G. (2007). Structure and function of human plasma

carboxypeptidase N, the anaphylatoxin inactivator. International

immunopharmacology 7, 1888-1899.

Starke, R.D., Paschalaki, K.E., Dyer, C.E., Harrison-Lavoie, K.J., Cutler, J.A.,

McKinnon, T.A., Millar, C.M., Cutler, D.F., Laffan, M.A., and Randi, A.M. (2013).

Cellular and molecular basis of von Willebrand disease: studies on blood

outgrowth endothelial cells. Blood 121, 2773-2784.

Stevenson, S., Mallett, A., Oliver, K., Hyland, V., Hawley, C., Malmanche, T., and

Isbel, N. (2014). Atypical HUS associated with severe, unexpected antibody-

Page 132: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

120

mediated rejection post kidney transplant. Nephrology (Carlton) 19 Suppl 1, 22-

26.

Symmers, W.S. (1952). Thrombotic microangiopathic haemolytic anaemia

(thrombotic microangiopathy). British medical journal 2, 897-903.

Tan, K., Lessieur, E., Cutler, A., Nerone, P., Vasanji, A., Asosingh, K., Erzurum, S.,

and Anand-Apte, B. (2010). Impaired function of circulating CD34(+) CD45(-)

cells in patients with proliferative diabetic retinopathy. Experimental eye

research 91, 229-237.

Tarr, P.I., Gordon, C.A., and Chandler, W.L. (2005). Shiga-toxin-producing

Escherichia coli and haemolytic uraemic syndrome. Lancet 365, 1073-1086.

Tati, R., Kristoffersson, A.C., Stahl, A.L., Rebetz, J., Wang, L., Licht, C., Motto, D., and

Karpman, D. (2013). Complement activation associated with ADAMTS13

deficiency in human and murine thrombotic microangiopathy. J Immunol 191,

2184-2193.

Thompson, R.A., and Winterborn, M.H. (1981). Hypocomplementaemia due to a

genetic deficiency of beta 1H globulin. Clinical and experimental immunology 46,

110-119.

Thurman, J.M., and Renner, B. (2011). Dynamic control of the complement

system by modulated expression of regulatory proteins. Laboratory

Investigation 91, 4-11.

Timmermans, F., Plum, J., Yoder, M.C., Ingram, D.A., Vandekerckhove, B., and

Case, J. (2009). Endothelial progenitor cells: identity defined? J Cell Mol Med 13,

87-102.

Timmermans, F., Van Hauwermeiren, F., De Smedt, M., Raedt, R., Plasschaert, F.,

De Buyzere, M.L., Gillebert, T.C., Plum, J., and Vandekerckhove, B. (2007).

Endothelial outgrowth cells are not derived from CD133+ cells or CD45+

hematopoietic precursors. Arterioscler Thromb Vasc Biol 27, 1572-1579.

Toshner, M., Dunmore, B.J., McKinney, E.F., Southwood, M., Caruso, P., Upton, P.D.,

Waters, J.P., Ormiston, M.L., Skepper, J.N., Nash, G., et al. (2014). Transcript

analysis reveals a specific HOX signature associated with positional identity of

human endothelial cells. PloS one 9, e91334.

Page 133: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

121

Tsai, H.M. (1996). Physiologic cleavage of von Willebrand factor by a plasma

protease is dependent on its conformation and requires calcium ion. Blood 87,

4235-4244.

Tsai, H.M. (2003). Is severe deficiency of ADAMTS-13 specific for thrombotic

thrombocytopenic purpura? Yes. J Thromb Haemost 1, 625-631.

Tsai, H.M. (2010). Pathophysiology of thrombotic thrombocytopenic purpura.

International journal of hematology 91, 1-19.

Tsai, H.M. (2013). Untying the knot of thrombotic thrombocytopenic purpura

and atypical hemolytic uremic syndrome. The American journal of medicine 126,

200-209.

Tsai, H.M., and Lian, E.C. (1998). Antibodies to von Willebrand factor-cleaving

protease in acute thrombotic thrombocytopenic purpura. The New England

journal of medicine 339, 1585-1594.

Tsai, H.M., Raoufi, M., Zhou, W., Guinto, E., Grafos, N., Ranzurmal, S., Greenfield,

R.S., and Rand, J.H. (2006). ADAMTS13-binding IgG are present in patients with

thrombotic thrombocytopenic purpura. Thrombosis and haemostasis 95, 886-

892.

Tschopp, J., and French, L.E. (1994). Clusterin: modulation of complement

function. Clinical and experimental immunology 97 Suppl 2, 11-14.

Tura, O., Skinner, E.M., Barclay, G.R., Samuel, K., Gallagher, R.C., Brittan, M.,

Hadoke, P.W., Newby, D.E., Turner, M.L., and Mills, N.L. (2013). Late outgrowth

endothelial cells resemble mature endothelial cells and are not derived from

bone marrow. Stem Cells 31, 338-348.

Turner, N., Nolasco, L., Dong, J.F., and Moake, J. (2009). ADAMTS-13 cleaves long

von Willebrand factor multimeric strings anchored to endothelial cells in the

absence of flow, platelets or conformation-altering chemicals. J Thromb Haemost

7, 229-232.

Turner, N., Nolasco, L., and Moake, J. (2012). Generation and breakdown of

soluble ultralarge von Willebrand factor multimers. Seminars in thrombosis and

hemostasis 38, 38-46.

Turner, N., Nolasco, L., Nolasco, J., Sartain, S., and Moake, J. (2014). Thrombotic

Microangiopathies and the Linkage between von Willebrand Factor and the

Page 134: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

122

Alternative Complement Pathway. Seminars in thrombosis and hemostasis 40,

544-550.

Turner, N.A., and Moake, J. (2013). Assembly and activation of alternative

complement components on endothelial cell-anchored ultra-large von

Willebrand factor links complement and hemostasis-thrombosis. PloS one 8,

e59372.

Turner, N.A., Walker, J.H., Ball, S.G., and Vaughan, P.F. (1996). Phorbol ester-

enhanced noradrenaline secretion correlates with the presence and activity of

protein kinase C-alpha in human SH-SY5Y neuroblastoma cells. J Neurochem 66,

2381-2389.

Valentijn, K.M., and Eikenboom, J. (2013). Weibel-Palade bodies: a window to

von Willebrand disease. J Thromb Haemost 11, 581-592.

Valentijn, K.M., Sadler, J.E., Valentijn, J.A., Voorberg, J., and Eikenboom, J. (2011).

Functional architecture of Weibel-Palade bodies. Blood 117, 5033-5043.

Van Hove, J.L., Van Damme-Lombaerts, R., Grunewald, S., Peters, H., Van Damme,

B., Fryns, J.P., Arnout, J., Wevers, R., Baumgartner, E.R., and Fowler, B. (2002).

Cobalamin disorder Cbl-C presenting with late-onset thrombotic

microangiopathy. Am J Med Genet 111, 195-201.

Wagner, S.J., and Myrup, A.C. (2005). Toward closed-system culture of blood

origin endothelial cells. Transfusion 45, 1201-1207.

Wallis, R. (2007). Interactions between mannose-binding lectin and MASPs

during complement activation by the lectin pathway. Immunobiology 212, 289-

299.

Walport, M.J. (2001a). Complement. First of two parts. The New England journal

of medicine 344, 1058-1066.

Walport, M.J. (2001b). Complement. Second of two parts. The New England

journal of medicine 344, 1140-1144.

Wang, J.W., Bouwens, E.A., Pintao, M.C., Voorberg, J., Safdar, H., Valentijn, K.M., de

Boer, H.C., Mertens, K., Reitsma, P.H., and Eikenboom, J. (2013a). Analysis of the

storage and secretion of von Willebrand factor in blood outgrowth endothelial

cells derived from patients with von Willebrand disease. Blood 121, 2762-2772.

Wang, J.W., Bouwens, E.A., Pintao, M.C., Voorberg, J., Safdar, H., Valentijn, K.M., de

Boer, H.C., Mertens, K., Reitsma, P.H., and Eikenboom, J.C. (2013b). Analysis of

Page 135: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

123

storage and secretion of von Willebrand factor in blood outgrowth endothelial

cells derived from von Willebrand disease patients. Blood.

Warwicker, P., Donne, R.L., Goodship, J.A., Goodship, T.H., Howie, A.J.,

Kumararatne, D.S., Thompson, R.A., and Taylor, C.M. (1999). Familial relapsing

haemolytic uraemic syndrome and complement factor H deficiency. Nephrol Dial

Transplant 14, 1229-1233.

Warwicker, P., Goodship, J.A., and Goodship, T.H. (1998a). Factor H--US? Nephrol

Dial Transplant 13, 1921-1923.

Warwicker, P., Goodship, T.H., Donne, R.L., Pirson, Y., Nicholls, A., Ward, R.M.,

Turnpenny, P., and Goodship, J.A. (1998b). Genetic studies into inherited and

sporadic hemolytic uremic syndrome. Kidney international 53, 836-844.

Waters, A.M., and Licht, C. (2011). aHUS caused by complement dysregulation:

new therapies on the horizon. Pediatr Nephrol 26, 41-57.

Wirstlein, P.K., Jasinski, P., Rajewski, M., Gozdziewicz, T., and Skrzypczak, J.

(2012). Complement inhibitory proteins expression in placentas of

thrombophilic women. Folia Histochemica et Cytobiologica 50, 460-467.

Wu, T.C., Yang, S., Haven, S., Holers, V.M., Lundberg, A.S., Wu, H., and Cataland,

S.R. (2013). Complement activation and mortality during an acute episode of

thrombotic thrombocytopenic purpura. J Thromb Haemost 11, 1925-1927.

Yang, B., Xi, X., Aronowski, J., and Savitz, S.I. (2012). Ischemic stroke may activate

bone marrow mononuclear cells to enhance recovery after stroke. Stem cells and

development 21, 3332-3340.

Yoon, C.H., Hur, J., Park, K.W., Kim, J.H., Lee, C.S., Oh, I.Y., Kim, T.Y., Cho, H.J., Kang,

H.J., Chae, I.H., et al. (2005). Synergistic neovascularization by mixed

transplantation of early endothelial progenitor cells and late outgrowth

endothelial cells: the role of angiogenic cytokines and matrix metalloproteinases.

Circulation 112, 1618-1627.

Zheng, X., Chung, D., Takayama, T.K., Majerus, E.M., Sadler, J.E., and Fujikawa, K.

(2001). Structure of von Willebrand factor-cleaving protease (ADAMTS13), a

metalloprotease involved in thrombotic thrombocytopenic purpura. The Journal

of biological chemistry 276, 41059-41063.

Page 136: Complement Regulation on Vascular Endothelial Cells ...€¦ · 2.3.8.2 BioFlux (Fluxion Biosciences) Microfluidic System ..... 35 2.4 RESULTS 37 2.4.1 BOEC Have An Endothelial Cell

124

Zipfel, P.F., Misselwitz, J., Licht, C., and Skerka, C. (2006). The role of defective

complement control in hemolytic uremic syndrome. Seminars in thrombosis and

hemostasis 32, 146-154.

Zipfel, P.F., and Skerka, C. (2009). Complement regulators and inhibitory

proteins. Nature reviews Immunology 9, 729-740.

Zipfel, P.F., Wolf, G., John, U., Kentouche, K., and Skerka, C. (2011). Novel

developments in thrombotic microangiopathies: is there a common link between

hemolytic uremic syndrome and thrombotic thrombocytic purpura? Pediatr

Nephrol 26, 1947-1956.