121
Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation of Complement Alternative Pathway by Joshua Yuen A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Laboratory Medicine & Pathobiology University of Toronto © Copyright by Joshua Yuen 2013

Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

  • Upload
    others

  • View
    16

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated

Activation of Complement Alternative Pathway

by

Joshua Yuen

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

Department of Laboratory Medicine & Pathobiology University of Toronto

© Copyright by Joshua Yuen 2013

Page 2: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

ii

Properdin Binds Pseudomonas aeruginosa and is Required for

Neutrophil Extracellular Trap Mediated Activation of Complement

Alternative Pathway

Joshua Yuen

Master of Science

Department of Laboratory Medicine & Pathobiology

University of Toronto

2013

Abstract

Neutrophils play an important, yet poorly understood role, in complement mediated pathologies.

Here we identified that neutrophils contain key components from the complement alternative

pathway: properdin (CFP), complement component 3 (C3), complement factor B (CFB), and

complement factor H (CFH). Activation of neutrophils resulted in secretion of these complement

components. When neutrophils are further activated to form neutrophil extracellular traps

(NETs), CFP is deposited onto the surfaces of the NETs. In addition, CFP is able to bind to

Pseudomonas aeruginosa, an opportunistic bacterium which can activate neutrophils to form

NETs. Furthermore, NETs activate complement and increase formation of the terminal

complement complex. The activation of complement on NETs can be initiated through multiple

pathways, however, activation of the alternative pathway is dependent on CFP. This mechanism,

potentially required for effective host defense, may also contribute to complement activation and

disease.

Page 3: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

iii

Acknowledgments

I would like to thank my supervisors, Dr. Christoph Licht and Dr. Nades Palaniyar for their

continued support and guidance. Their passion for research was inspiring and helped me become

a better researcher and critical thinker. More importantly, they were determined to guide me

along the way as I developed personal skills that would help me not only succeed academically,

but also succeed in life. For that I will forever be thankful.

To my Master’s advisory committee, Dr. Walter Kahr and Dr. Margaret Rand, you have been

very supportive and encouraging. Thank you.

I would also like to thank my colleagues in both labs for their continuous help and support. It has

been a true privilege to work with such an intelligent group of scientists. They really helped

motivate me to push forward when times were tough. I would especially like to thank David

Douda for all of his help, from optimizing results and experimental procedures to helpful

discussions and technical help. This work has been a collection of our blood, sweat and tears.

I would also like to thank Damien Noon, M.D., Hailu Huang, M.D., Hong Wang, M.D., and Jalil

Nasiri for their technical assistance in collecting peripheral blood.

A special thanks goes out to all the donors who contributed to this research. None of this work

would have been possible without their help.

I would like to thank Dr. Marina Ulanova from the Northern Ontario School of Medicine for

providing the GFP expressing Pseudomonas aeruginosa.

Finally, I would like to thank my parents for their unconditional love and support.

This work was supported by a University of Toronto Fellowship and an Ontario Graduate

Scholarship.

Page 4: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

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

List of Tables ............................................................................................................................... viii

List of Figures ................................................................................................................................ ix

List of Appendices ......................................................................................................................... xi

List of Abbreviations .................................................................................................................... xii

1 Introduction ............................................................................................................................ 1

1.1 The Complement System ....................................................................................................... 1

1.1.1 Classical Pathway ........................................................................................................ 1

1.1.2 Lectin Pathway ............................................................................................................ 2

1.1.3 Alternative Pathway .................................................................................................... 2

1.1.4 Amplification and Terminal Pathway ......................................................................... 4

1.1.5 Other Activation Pathways .......................................................................................... 6

1.2 Complement in Disease ......................................................................................................... 6

1.2.1 Endogenous Activation ............................................................................................... 7

1.2.2 Complement Dysregulation ......................................................................................... 8

1.3 Neutrophils ............................................................................................................................. 9

1.3.1 Neutrophil Activation and Resolution ......................................................................... 9

1.3.2 Neutrophil Extracellular Traps .................................................................................. 12

1.3.3 Neutrophil Extracellular Traps in Disease ................................................................ 15

1.4 Complement and NETs in Disease ...................................................................................... 16

1.5 Complement and Neutrophils .............................................................................................. 22

1.6 Neutrophil Complement Cross-talk .................................................................................... 23

1.7 Rationale .............................................................................................................................. 25

Page 5: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

v

1.8 Hypothesis ............................................................................................................................ 27

1.9 Aims ..................................................................................................................................... 27

2 Methods ................................................................................................................................ 28

2.1 Reagents ............................................................................................................................... 28

2.2 Research Ethics Board Approval ......................................................................................... 28

2.3 Neutrophil Isolation ............................................................................................................. 28

2.4 Preparation of Neutrophil Lysates ....................................................................................... 28

2.5 Bacterial Cultures and Strains .............................................................................................. 29

2.5.1 Bacterial Plating ........................................................................................................ 29

2.5.2 Bacterial Growth ....................................................................................................... 29

2.6 Neutrophil Activation .......................................................................................................... 30

2.6.1 Neutrophil Oxidative Burst ....................................................................................... 30

2.6.2 Neutrophil Activation and Secretion ......................................................................... 30

2.6.3 Differential Cell Counts for Nuclear Morphology .................................................... 30

2.6.4 SYTOX Green Plate Reader Assay ........................................................................... 31

2.7 Western Blot ........................................................................................................................ 31

2.7.1 Determining Protein Concentration ........................................................................... 32

2.8 Imaging and Microscopy ..................................................................................................... 32

2.8.1 Acid Washed Cover Slips .......................................................................................... 32

2.8.2 Preparation of Cover Slips for Microscopy ............................................................... 33

2.8.3 Seeding Neutrophils on Coated Cover Slips ............................................................. 33

2.9 Identifying Complement Proteins in Neutrophils ................................................................ 33

2.10 Detection of Complement Proteins in NETs ....................................................................... 34

2.11 Complement Competent Plasma .......................................................................................... 34

2.12 Preparation of Rabbit Erythrocytes ...................................................................................... 35

2.13 Plasma Complement Activity Assay ................................................................................... 35

Page 6: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

vi

2.14 Complement Activation and C5b-9 Formation on NETs .................................................... 35

2.15 Inhibiting Properdin Activity in NETs ................................................................................ 36

2.16 Spinning-Disc Confocal Microscopy ................................................................................... 37

2.17 Colocalization Analysis ....................................................................................................... 37

2.18 Statistical Analysis ............................................................................................................... 37

3 Results – Aim 1 .................................................................................................................... 38

3.1 Neutrophils Contain Complement Factors ........................................................................... 38

3.2 Investigating Neutrophil Activation .................................................................................... 41

3.2.1 Neutrophil Activation and Oxidative Burst ............................................................... 41

3.2.2 Complement Protein Localization Upon Activation ................................................. 43

3.2.3 Complement Protein Secretion in Response to Activation ....................................... 45

3.3 Quantification of Nuclear Morphologies During NETosis .................................................. 48

3.4 PMA, but not C5a and fMLP, Induce NET Formation ........................................................ 50

3.5 Neutrophil Extracellular Traps Contain Properdin .............................................................. 52

3.6 NETs Increase Formation of Terminal Complement Complex ........................................... 57

3.7 NETs Activate Complement Alternative Pathway to Form C5b-9 ..................................... 59

3.8 NETs Activate Complement and Activation of Complement AP on NETs is Dependent

on Properdin ......................................................................................................................... 61

4 Results – Aim 2 .................................................................................................................... 64

4.1 Identifying Bacteria That Induce NETosis .......................................................................... 64

4.2 Neutrophil Properdin Binds P. aeruginosa Independent of NETs ...................................... 68

4.3 NETs Increase Formation of C5b-9 in Response to Bacteria .............................................. 70

4.4 NETs Activate Complement Alternative Pathway in Response to Bacteria ....................... 72

4.5 NETs Activate Complement in Response to P. aeruginosa and Complement AP

Activation is Dependent on Properdin ................................................................................. 74

5 Discussion ............................................................................................................................ 76

5.1 Characterizing Interactions of Complement and Neutrophils ............................................. 76

Page 7: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

vii

5.2 Properdin is Found on Neutrophil Extracellular Traps ........................................................ 78

5.3 Neutrophil Extracellular Traps Activate Complement ........................................................ 79

5.4 Neutrophils form NETs in Response to Bacteria ................................................................. 80

5.5 NETs Formed in Response to Bacteria Activate Complement ............................................ 80

6 Conclusion and Future Directions ....................................................................................... 84

6.1 Conclusions .......................................................................................................................... 84

6.2 Future Directions ................................................................................................................. 84

7 References ............................................................................................................................ 87

8 Appendix ............................................................................................................................ 103

Page 8: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

viii

List of Tables

Table 1. Activators of NETs ......................................................................................................... 14

Page 9: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

ix

List of Figures

Figure 1. Complement: Key mediator of innate immunity. ............................................................ 5

Figure 2. Neutrophil activation and regulation. ............................................................................ 11

Figure 3. Complement and NETs lead to endothelial cell damage. .............................................. 17

Figure 4. Complement interaction with NETs. ............................................................................. 26

Figure 5. Neutrophils contain proteins from complement alternative pathway. ........................... 40

Figure 6. Neutrophil stimulation with fMLP and PMA, but not C5a, leads to oxidative burst. ... 42

Figure 7. Complement proteins associate with sialic acid upon activation. ................................. 44

Figure 8. Activated neutrophils secrete complement proteins. ..................................................... 47

Figure 9. Activated neutrophils form neutrophil extracellular traps. ............................................ 49

Figure 10. PMA, but not C5a and fMLP, induces NET formation. .............................................. 51

Figure 11. Neutrophil extracellular traps contain properdin. ........................................................ 53

Figure 12. C3 does not interact with neutrophil extracellular traps. ............................................. 54

Figure 13. CFB does not interact with neutrophil extracellular traps. .......................................... 55

Figure 14. CFH does not interact with neutrophil extracellular traps. .......................................... 56

Figure 15. Neutrophil extracellular traps increase formation of C5b-9. ....................................... 58

Figure 16. NETs activate complement alternative pathway to form C5b-9 ................................. 60

Figure 17. Properdin is required for activation of complement AP on NETs. ............................. 63

Figure 18. Pseudomonas aeruginosa activate neutrophils to form NETs .................................... 66

Figure 19. Neutrophils activated with S. aureus, E. coli, and B. subtilis. .................................... 67

Page 10: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

x

Figure 20. Properdin binds P. aeruginosa independent of NETs. ................................................ 69

Figure 21. P. aeruginosa activation of neutrophils increases C5b-9 formation on NETs. ........... 71

Figure 22. NETs activate complement AP to form C5b-9 in presence of P. aeruginosa. ............ 73

Figure 23. NETs activation of complement AP in presence of P. aeruginosa is dependent on

Properdin. ...................................................................................................................................... 75

Figure 24. Proposed model of complement-neutrophil interactions. ............................................ 83

Page 11: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

xi

List of Appendices

Appendix A. Optimizing the Protocol for Neutrophil Lysates ................................................... 103

Appendix B. Hemolysis Assay Data ........................................................................................... 106

Page 12: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

xii

List of Abbreviations

aHUS atypical Hemolytic Uremic Syndrome

ALT alanine aminotransferase

ACPA anti-citrullinated peptide antibody

ANCA anti- neutrophil cytoplasmic antibody

AP alternative pathway

BCA bicinchoninic acid

C1qR C1q receptor

C3aR C3a receptor

C5aR C5a receptor

CD59 membrane attack complex inhibitor

CFB complement factor B

CFD complement factor D

CFH complement factor H

CFHR1 complement factor H related protein 1

CFI complement factor I

CFP properdin or complement factor P

CGD chronic granulomatous disease

CP classical pathway

CR1 complement receptor 1 (also CD35)

CR3 complement receptor 3 (also Mac-1 or CD11b/CD18)

CR4 complement receptor 4 (CD11c/CD18)

Page 13: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

xiii

DAF decay accelerating factor (also CD55)

DAMP damage associated molecular pattern

DPI diphenyleneiodonium

FFP fresh frozen plasma

fMLP n-formyl-methyl-leucyl-phenylalanine

GAGs glycosaminoglycans

G-CSF granulocyte colony stimulating factor

GFP green fluorescent protein

GM-CSF granulocyte-macrophage colony stimulating factor

HUS Hemolytic Uremic Syndrome

HUVEC human umbilical vein endothelial cell

ICAM-1 Intercellular adhesion molecule 1

IL-1β interleukin-1 beta

IL-8 interleukin 8

IL-17 interleukin 17

IL-23 interleukin 23

LPS lipopolysaccharide

LP lectin pathway

MASP MBL associated serine protease

MBL mannose binding lectins

MCP membrane cofactor protein

MOI multiplicity of infection

Page 14: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

xiv

MPO myeloperoxidase

NE neutrophil elastase

NETs neutrophil extracellular traps

NOX NADPH oxidase

PAD4 peptidylarginine deiminase 4

PAMP pathogen associated molecular pattern

PKC protein kinase-C

PMA phorbol 12-myristate 13-acetate

PR3 proteinase 3

PSGL-1 p-selectin glycoprotein ligand-1

ROS reactive oxygen species

SLE systemic lupus erythematosus

STX shiga toxin

SVV small vessel vasculitis

TCC terminal complement complex (C5b-9)

TNF-α tumor necrosis factor-α

TLR4 toll-like receptor 4

TSR thrombospondin repeats

Page 15: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

1

1 Introduction

1.1 The Complement System

The complement system represents one of the most ancient cornerstones of innate immunity1. It

consists of over 40 soluble and membrane bound proteins2,3

. This highly integrated network,

consisting of many signaling molecules and regulators, acts as an intricate immune surveillance

system to discriminate between healthy host tissue, unwanted cellular debris, apoptotic cells,

necrotic cells and microbial invaders4.

Traditionally, activation of the complement system is achieved through three distinct

initiation pathways: classical (CP), lectin (LP), and alternative (AP). These cascade-like

pathways all culminate to the formation of the convertases, the major enzyme of complement

activation5,6

, which converge to a common effector C3, generating opsonin C3b and

anaphylatoxin C3a. Further activation leads to the formation of C5 convertases which direct the

assembly of the terminal complement complex (C5b-9), a lytic pore which assembles on

membranes to mediate cell destruction5,6

. More recently, a new proteolytic pathway in which

thrombin acts as a C5 convertase, cleaving C5 to the anaphylatoxin C5a, has been described7.

1.1.1 Classical Pathway

The classical pathway is initiated by the calcium dependent C1 complex through its pattern

recognition molecule C1q. Classical pathway is strongly initiated through binding of C1q to IgG

or IgM clusters8. As such, it is often referred to as the antibody-dependent pathway. However,

C1q is also capable of recognizing distinct structures directly on microbial cells9. In addition to

endogenous pattern recognition molecules (immunoglobulins, C-reactive protein), C1q can also

bind to prions, DNA, late apoptotic, and necrotic cells10

. C1q binding leads to conformational

changes in the C1 complex allowing interaction between the C1r and C1s proteases11

. C1r

undergoes autoproteolytic activation cleaving the zymogen of C1s. C1s is a highly specific

protease which cleaves C4 into C4a and C4b, and C2 into C2a and C2b to form the classical

pathway C3 convertase (C4b2b). C4b also opsonizes targets further directing C3 convertase

assmembly on membrane surfaces. This convertase can cleave C3 to initiate amplification and

downstream functions of complement amplification.

Page 16: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

2

1.1.2 Lectin Pathway

The lectin pathway is initiated through recognition of carbohydrates on microbial surfaces by

Mannose-binding lectin (MBL) or ficolin. MBL belong to the collectin family of proteins which

contain a carbohydrate recognition domain and a collagen-like domain12

. The carbohydrate

recognition domain of MBL binds to carbohydrates with 3- and 4- hydroxyl groups in the

pyranose ring in a calcium dependent manner13

. This allows for effective recognition of

carbohydrate surfaces (e.g. N-acetyl-glucosamine [GlcNAc]) on pathogens and leaves

mammalian glycoproteins (e.g. galactose and sialic acid), which do not fit this steric

requirement, virtually undetected 14

. Ficolins, like MBL, contain a collagen-like stem structure,

however they also contain a fibrinogen-like domain. Serum ficolin are lectins that have similar

binding specificity as MBL.

MBL-associated serine proteases (MASP) are a serine-protease superfamily (MASP1,

MASP2, sMAP/MAp19, MASP3). Structurally, they are similar to the proteolytic components

(C1r/C1s) of the classical pathway15,16

. Binding of MBL or ficolins to carbohydrates activates

lectin-pathway MASP through mechanisms that are currently unknown17

. MASP2, like C1s,

cleaves C4 and C2 to form the C3 convertase (C3b2b), which is common to the CP C3

convertase16

. In contrast, MASP1 can cleave C2 but is unable to cleave C4, suggesting that it can

enhance the activation of lectin pathway triggered by MBL-MASP2 complexes but not initiate

lectin pathway itself18

.

1.1.3 Alternative Pathway

In contrast to the inducible lectin and classical, the alternative pathway (AP) is constitutively

active and interacts with cell surfaces for constant immune surveillance. Initiation of AP begins

in the fluid phase where C3 is hydrolyzed to C3H2O, exposing new binding sites to allow for rapid

probing of cells in a process commonly referred to as the tick-over pathway19

. Complement

factor B (CFB) protease binds to C3H2O and is cleaved by complement factor D (CFD), in the

presence of Mg2+

, releasing the complement factor B amino terminal fragment (Ba) and

activating the serine protease domain (Bb), to form fluid phase C3 convertase (C3H2OBb)20

.

Fluid phase C3H2OBb cleaves C3 into C3a and C3b. C3b binds to amine and carbohydrate

groups on cell surfaces and engages CFB. Once again, CFB is cleaved by CFD, in the presence

of Mg2+

, to form the AP C3 convertase (C3bBb). The half-life of the C3bBb complex is

Page 17: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

3

shortlived (T1/2 ~ 90 sec21,22

) but is stabilized 5 to 10 fold by association with properdin (CFP) to

form a stable C3 convertase (C3bBbP)23

.

On foreign particles, unrestricted activation of AP can lead to rapid amplification of the

complement cascades. This process, however, is immediately regulated on host cells by

regulatory factors. These regulatory factors include complement factor H (CFH), complement

factor I (CFI), and membrane bound: decay accelerating factor (DAF), membrane cofactor

protein (MCP), complement receptor 1 (CR1/CD35) and membrane attack complex inhibitor

(CD59). CFH is the major regulator of the AP24

. CFH regulates AP by either, competitively

binding to C3b preventing the formation of the C3bBb complex, or serving as a cofactor for CFI

mediated cleavage of C3b25,26

. Thus transient formation and regulation of complement AP C3

convertase is in dynamic equilibrium.

In addition to stabilizing the complement AP C3 convertase, CFP also serves as a pattern

recognition molecule to initiate activation of complement AP. Recent studies highlight the

emerging roles for CFP to act as a pattern recognition molecule capable of recognizing pathogen

or damage associated molecular patterns (DAMP/PAMP)27

. Using surface plasmon resonance

methodology, Spitzer et al, demonstrated that CFP binding provides a platform for convertase

assembly28

. CFP has been shown to bind to glycosaminoglycan, Neisseria gonorrhoeae, (LPS)-

defective Escherichia coli, LPS, early/late apoptotic T cells, and necrotic cells via DNA

surfaces28-32

.

Properdin is a 53 kDa protein composed of 7 thrombospondin repeats (TSR)33,34. The TSR

repeats are numbered from 0-6 (TSR 0-6) with TSR 0 starting at the N-terminus35. It is a rod-like

protein, approximately 26 nm in length and 2.6 nm in diameter36. In circulation, CFP is a multimeric

protein that forms dimers, trimers and tetramers in a head to tail fashion33. CFP functions are

dependent on its polymeric nature as the activity of CFP increases with size. The CFP tetramer is

approximately 10 fold more active when compared to the dimer34. The detailed three-dimensional

structure of CFP has not yet been resolved, however a protein surface on TSR 2 and 3 of CFP

containing alternating arginine and tryptophan side chains has been proposed as the site for pattern

recognition37. The distance between the arginines (~9 Å) closely matches the length of a

glycosaminoglycan disaccharide unit. This suggests that interactions between CFP and

glycosaminoglycan are determined through electrostatic interactions33

. The CFP residues that are

proposed to mediate binding to sulfate moieties of glycosaminoglycan may also mediate

Page 18: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

4

interactions with other large polyanionic molecules. Because properdin can interact with a

variety of polyanionic macromolecules, any properdin-opsonized target can potentially amplify

complement23

.

1.1.4 Amplification and Terminal Pathway

All surface bound C3 convertase, regardless of origin, induces amplification of AP by cleaving

C3 to deposit C3b in the vicinity of complement activation. In the presence of CFB and CFD,

this allows for an efficient cycle of C3 cleavage and AP convertase assembly. Despite its name,

the alternative pathway contributes to 80-90% of the final C5a generation, regardless of the

initial triggering cascade38

. Amplification of complement by AP causes a rapid increase in C3

cleavage and C3b deposition onto surfaces. Deposition of C3b onto C3 convertases drives the

formation of the C5 convertases (C4b2b3b/C3bBb3b) shifting substrate specificity from C3 to

C539

. C5 is cleaved into C5a, a powerful anaphylatoxin, and C5b, which directs a non enzymatic

assembly of the terminal pathway components C6, C7, C8, and C9 (C5b-9) to form the cell lytic

terminal complement complex (TCC)40

. Thus, activation of complement through its various

initiating pathways can trigger a cascade which leading to the formation of C5b-9 (Figure 1).

Page 19: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

5

Figure 1. Complement: Key mediator of innate immunity.

Complement activation can be initiated through three major pathways. The alternative pathway

is constitutively active, while the classical and lectin pathways are inducible. In addition to these

three pathways, serine proteases can directly cleave complement proteins to initiate complement

activity. Complement activation occurs in a sequential manner. All three pathways converge to

form an enzyme known as the C3 convertase. (C3bBb for the alternative pathway and C4bC2b

for the classical and lectin pathway). The C3 convertases cleave C3 to produce anaphylatoxin

C3a and opsonin C3b. Continued activation and deposition of C3b onto cell surfaces leads to the

formation of complement C5 Convertase (C3bBbC3b for the alternative pathway and

C4bC2bC3b for the classical and lectin pathways). C5 convertases cleave C5 into anaphylatoxin

C5a, and generates C5b which initiates the assembly of the terminal complement complex (C5b-

9). Reprinted by permission from Macmillan Publishers Ltd: Nature Reviews Immunology 9,

729-740, 2009.

Page 20: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

6

1.1.5 Other Activation Pathways

In addition to the three traditional pathways of complement activation, direct cleavage of C3 or

C5 can also occur through a number of extrinsic proteases. In an in vitro assay, Vogt

demonstrated that leukocyte elastase can cleave C5 to form biologically active C5b-6 like

complex to initiate formation of TCC41

. Using a C3-/-

mice, Huber-Lang et al, demonstrated that

thrombin can cleave C5 to produce biologically active C5a/C5b in the absence of C5

convertase7. Other proteases that have been known to cleave complement C3/C5 include plasmin

and plasma kallikrein2. These extrinsic pathways for complement activation suggest that there is

cross talk between complement, innate immune cells and coagulation.

1.2 Complement in Disease

The primary task of the complement system is to protect the host against invading microbes.

Sensing various danger signals, both host derived and foreign, complement is able to execute its

function in a tightly regulated manner, thus mediating the inflammatory response3. For some

types of danger signals, the complement system serves as an opsonin, tagging molecules for

clearance, where as for others it can mediate a strong inflammatory response42

.

The inflammatory response is a pathophysiological process that engages hundreds of

mediators, cell types, and tissues. Inflammation is often thought of as a response to infection,

however, it can be initiated by any cell stimulus causing injury, including chemical or physical

injury43,44

. The inflammatory process eliminates the initiating factor with minimal destruction to

host tissues. Central to inflammation, complement is intricately involved in regulating this

process4. The mechanisms for complement activation, thus regulation of inflammation, are well

understood in the vasculature. However, soluble components can also infiltrate tissues and

organs to engage exogenous (e.g. infection) and endogenous (e.g. autoimmunity) stimuli that

could cause cell injury45

. Complement activation, under the tight control of inhibitors, can illicit

an appropriate immune response. However, any trigger that tips the delicate balance between

activation and regulation can potentially lead to pathology4,46,47

. There is a paradoxical role for

complement participation in inflammatory disorders. An overly aggressive complement system

results in inflammation-mediated tissue damage, however a deficiency or lack of complement

response leaves the host immune compromised4.

Page 21: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

7

The innate immune requirement for complement has been studied for decades but the

appreciation for its link to disease has only emerged in recent years48

. While we recognize that

complement deficiency leads to immune compromised individuals, the focus of our study is on

complement activation and inflammatory damage. Complement activation, through different

intitation pathways, produces pro-inflammatory peptides that mediate disease processes49

.

Deleterious complement activation precipitating into pathology is mediated through two major

mechanisms: Endogenous activation of complement by foreign trigger overwhelms regulatory

machinery to precipitate disease, or insufficient regulation of complement activation via

mutation, decreased expression of regulatory proteins or auto-antibodies leads to increased

complement activation50-52

.

1.2.1 Endogenous Activation

Hemolytic uremic syndrome (HUS) is a kidney disease characterized by microangiopathic

hemolytic anemia, thrombocytopenia and renal impairment. This disease is most commonly

triggered by shiga-like toxin (Stx)-producing E. coli and manifests with diarrhea, often bloody53

.

In North American and Western Europe, 70% of HUS is secondary to infection with E. coli

serotype O157:H753-57

. Retrograde transport of Stx by endothelial cells enzymatically blocks

protein synthesis and inhibits expression of anti apoptotic proteins58,59

. This was traditionally

viewed as the mechanism for mediating cell damage and pathology. However, recent evidence

indicates complement activation also contributes to disease. Stx was demonstrated to activate

complement and bind to the major soluble regulator of complement AP, CFH60

. These data were

further corroborated in a clinical setting as patients with HUS were described to have elevated

levels of complement activation products Bb and SC5b-961

. Furthermore, the use of C5-blocker,

eculizumab, was able to successfully treat a three year old patient suffering from severe Stx

associated HUS62

.

In addition, the circulating levels of CFP in plasma are relatively low (4-6 μg/ml)63

. The

release of CFP from infiltrating neutrophils is a major determinant for complement AP

activation64

. In HUS, neutrophils have been shown to mediate endothelial cell death, and patients

with high peripheral counts of neutrophils have poor prognosis65

. An influx in neutrophils will

lead to localized increase in complement AP activation. Whether the activation of complement

AP by infiltrating neutrophils contributes to pathology has yet to be determined.

Page 22: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

8

1.2.2 Complement Dysregulation

Atypical hemolytic uremic syndrom (aHUS) is a rare kidney disease characterized by

microangiopathic hemolytic anemia, thrombocytopenia and renal impairment. This disease

manifests without bloody diarrhea. Genetic analysis has linked aHUS with mutations in genes

encoding for the proteins of complement AP. Gain of function mutations to C366-68

and CFB69,70

and loss of function mutations to CFH71-73

, MCP9,10

, and CFI9,10

have all been described to

contribute to aHUS pathology. Moreover, the presence of anti CFH antibodies may also

precipitate aHUS pathology74,75

. During the onset of disease progression, patients have reduced

levels of circulating C3 and CFB, as well as increased levels of C3 activation fragments.

However, C4 is unaffected, suggesting that this pathology is dependent exclusively on AP76

.

Despite progress in identifying causative factors for aHUS pathogenesis, much of the

disease is enclosed in a shroud of mystery. While genetic mutation has been linked with

pathology, incidence among carriers with predisposing mutations counts for less than 50%76

. In

addition, while aHUS typically presents in childhood, the age of onset varies. Furthermore, the

variable expression of aHUS associated mutations makes it impossible to define a single factor

which may precipitate pathology. Taken together, these findings demonstrate that many other

factors may contribute to complement driven pathologies.

In 2011, a series of reports highlighted the potential or prominence for an infectious

trigger leading to aHUS77-80

. Currently there have been no studies to identify the link between

infection and aHUS pathogenesis. The clinical onset in a majority of familial aHUS cases occurs

subsequent to infection (bacterial or viral)81,82

. Therefore, neutrophils may be considered relevant

as they are the primary sentinel for immune surveillance in the blood stream. Neutrophils have

been shown to be able to directly activate complement. Infiltrating neutrophils can increase local

concentrations of complement CFP, thus activating complement AP83

. Furthermore, proteolytic

enzymes released from activated neutrophils (e.g. elastase) can cleave and activate C3 and C541

.

Neither of these concepts have been investigated within the context of NETs. The propensity for

neutrophil or possibly NETs (Section 1.3.2) to activate complement suggests that they may

trigger clinical manifestations of aHUS, however this has yet to be determined. Thus, we set

forth to investigate the interactions between neutrophils and complement.

Page 23: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

9

1.3 Neutrophils

Neutrophil granulocytes, also known as polymorphonuclear leukocytes, are the most abundant

leukocyte in the blood stream. They are highly motile and migrate to sites of infection or

inflammation through chemotaxis. As such, they are commonly referred to as the first line of

defense against infection84

.

They originate in the bone marrow, where under the influence of growth factors and

cytokines, pluripotent hematopoetic cells differentiate to form myeloblasts in a process called

myelopoesis. The cytokine granulocyte colony stimulating factor (G-CSF) is essential for

regulating neutrophil production in response to infection as G-CSF null mice develop chronic

neutropenia85

. The myeloblast progenitors, now committed to become granulocytes, synthesize

proteins and package them into different granules. The formation of granules is a continuous

process in which vesicles bud from the golgi apparatus and fuse forming granular structures.

During the course of maturation from the myeloblast, neutrophils alter their transcription profile

to synthesize various proteins86

. As a result, neutrophils form a continuum of granules with

overlapping contents87

. This is in stark contrast to the traditional method for the classification of

granules into azurophillic (primary), specific (secondary), and gelatinase (tertiary), based on their

protein constituents. While this division is practical, it is now appreciated as largely artificial88-90

.

Unlike the first three classes of granules which are formed from the golgi apparatus,

secretory vesicles are formed during the final stage of neutrophil maturation through endocytosis

in the bone marrow91

. Secretory vesicles are reservoirs for a variety of membrane receptors and

adhesion molecules. They play an important role in mediating the early response for neutrophil

activation. As they are endocytotic in origin, mobilization of secretory vesicles does not result in

the release of proteolytic enzymes, only plasma proteins that form the matrix of secretory

vesicles92

.

1.3.1 Neutrophil Activation and Resolution

Neutrophils are key effectors of innate immune and inflammatory responses93

. As the primary

sentinel for immune surveillance, neutrophils traverse the blood stream continuously and

randomly probing the endothelium of the vessel walls94

. At sites of tissue injury or infection,

stimulants such as bacterial derived lipopolysaccharides (LPS) and N-Formyl-Methyl-Leucyl-

Page 24: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

10

Phenylalanine (fMLP), as well as host derived cytokines (e.g. TNF-α, IL-1β, IL-17) activate

endothelial cells to increase the expression of the adhesion molecules P-selectin, E-selectin, and

several members from the integrin superfamily, the ICAMs95

. This process coordinates the

adhesion of neutrophils, which constitutively express P-selectin glycoprotein ligand-1 (PSGL-1)

and L-selectin, to activated endothelium which express E-selectin96

. The selectin mediated

adhesion enables neutrophils to undergo their characteristic rolling, tethering, and

transendothelial cell migration directed by selectins, cytokines and chemokines97

. As neutrophils

travel through the interstitium, they are guided by chemotactic gradients from both host derived

chemokines (e.g. IL-8) and pathogen associated molecular patterns (PAMPs) to sites of infection

and inflammation98

. All the while, these chemoattractants bind to their respective neutrophil

receptors (often G-protein coupled receptors) and initiate the downstream signaling cascade

dominated by the MAPK/ERK pathway in preparation for neutrophil activation99

. Upon arrival

to sites of high chemoattractant concentration, neutrophils coordinate a strict regiment to deal

with potential threats through programmed phagocytosis, degranulation (releasing stored

antimicrobial contents), and NETosis (process of forming Neutrophil Extracellular Traps [NETs

section 1.3.2])100

.

Neutrophil activation is effective when dealing with foreign invaders, however,

deployment of lethal cargo of neutrophils results in collateral damage to host tissue101,102

. Thus,

neutrophils must be swiftly removed from sites of activation before they have detrimental effects

on host tissue. Resolution of inflammation removes apoptotic neutrophils from sites of

inflammation and prevents infiltration of additional cells. This is achieved through two major

mechanisms: phagocytosis and lipid mediators. The phagocytosis of apoptotic neutrophils by

macrophages results in down regulation of IL-23, a cytokine that controls IL-17 production by T-

cells103

. The reduction of IL-17 results in decreased production of G-CSF thus reducing

neutrophil production (Figure 2). In the later stages of the inflammatory response, neutrophils

interact with cells at sites of inflammation to produce anti-inflammatory lipid-mediators (e.g.

lipoxins, resolvins, protectins)104

. These lipid-mediators enhance uptake of apoptotic cells by

macrophages and down regulate neutrophil activation105,106

. The mechanisms for clearance of

NETs are currently unknown, however, DNase has been suggested to play an important role in

NET clearance. DNase impairment has been implicated in NET mediated pathology (see section

on Lupus Nephritis in Section 1.4).

Page 25: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

11

Figure 2. Neutrophil activation and regulation.

Neutrophils circulate the blood stream continuously and randomly probing endothelium.

Activation of endothelium results in increased expression of adhesion molecules (P-selectin and

E-Selectin) which capture neutrophils which express PSGL-1 and L-selectin to mediate rolling,

expression of neutrophil integrins and trans-endothelial cell migration. Neutrophils migrate to

sites of infection or inflammation through chemotaxis. Upon arrival to sites of high

chemoattractent concentration, neutrophils respond through generation of cytokines,

phagocytosis, degranulation or NETosis. Resolution of the inflammatory response is achieved

through phagocytosis of apoptotic neutrophils by macrophages. Reprinted from Immunity, 33,

Borregaard, N., Neutrophils, from Marrow to Microbes, 657-670, 2010, with permission from

Elsevier

Page 26: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

12

1.3.2 Neutrophil Extracellular Traps

In recent years, a unique form for neutrophil cell death, called NETosis, has been identified

whereby neutrophils actively release decondensed chromatin into extracellular space100,107

. These

structures, termed neutrophil extracellular traps (NETs), contain DNA, histones, granular and

cytoplasmic proteins100

. NETs can ensnare foreign invaders and confer antimicrobial activity by

exposing them to high local concentrations of antimicrobials108

. Mass spectrometry has identified

24 neutrophil proteins associated with NETs109

. This finding reveals that NET proteins are

primarily cationic as they interact with the polyanionic backbone of DNA. Bacteriacidal proteins

include: histones, defensins, elastase and myeloperoxidase, however any pattern recognition

molecule with high affinity for polyanionic compounds can potentially bind to NETs.

The mechanism for NET formation is not completely understood although

decondensation of chromatin is central to NETosis. This can be associated with the

hypercitrullination of histone H3 through peptidylarginine deiminase 4 (PAD4) which converts

arginine to citrulline, thus disrupting the charge interaction between positively charged arginine

and polyanionic DNA110

. PAD4 deficient mice are unable to produce NETs, although this has yet

to be confirmed ex vivo with primary human neutrophils111

. In addition, neutrophil elastase

infiltration from azurophil granules into the nucleus can degrade histones, driving chromatin

decondensation112

.

NET formation is dependent on production of reactive oxygen species (ROS). Hydrogen

peroxide generated by NADPH oxidase (NOX) is further metabolized by MPO. Inhibition of

NADPH oxidase with DPI prevents the formation of NETs. Furthermore, neutrophils from

patients with chronic granulomatous disease (CGD) and MPO deficient neutrophils are unable to

form NETs113,114

. ROS however is not sufficient for NET formation as healthy newborns are

unable to form NETs, despite being capable of full respiratory burst activity115

. Upstream of

NADPH oxidase, the Raf-MEK-ERK axis has been implicated in NET formation. Addition of

U0126, a specific MEK inhibitor, and ERK inhibitory peptide, a specific ERK2 inhibitor,

inhibited the formation of NETs, thus illustrating that the canonical Raf-MEK-ERK pathway is

involved in NETosis116

. In addition, the use of these inhibitors block the production of ROS,

implying that this pathway is upstream of NADPH oxidase116

. Independent of ROS production,

autophagy has also been shown to be required for NET formation. Pharmacological inhibition of

Page 27: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

13

PI3K with wortmannin prevents massive vacuolization of neutrophils to form NETs. Inhibition

of either ROS production (pharmacological or genetic) or neutrophil autophagy

(pharmacological) leads to apoptosis117

.

Currently, no surface receptor has been identified to directly induce the formation of

NETs, however, toll-like receptor 4 (TLR4) has been implicated to play an important role118,119

.

NETs can be formed in response to bacteria, protozoa, fungi, virus, host derived factors and

chemical compounds120

. With the exception of S. aureus, all known activators of NETs require

ROS production121

. For a complete summary of all activators of NETs known to date, please

refer to (Table 1).

Page 28: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

14

Table 1. Activators of NETs

NETosis Inducer In vitro In vivo Reference(s)

BACTERIA

Eschericia coli 10 MOI; bovine

0.01 MOI; human

-

-

115,118,122

Pseudomonas aeruginosa 0.1-10 MOI; human 1×106 CFU/mouse

123,124

Staphylococcus aureus 0.01-10 MOI; human - 100,121

Shigella flexneri 0.01 MOI; human 2.5-3.0×1010

/rabbit 100

Salmonella enterica 0.01 MOI; human - 100

Group A Streptococcus 0.1 MOI; human 5×107-2×10

8

CFU/mouse

125,126

Streptococcus pneumoniae 0.01 MOI; human 1×107/mouse

127

Mycobacterium tuberculosis 0.1-10 MOI; human - 128

PROTOZOA

Leishmania amazonensis 10 MOI; human - 129

Leishmania donovani 10 MOI; human - 130

Toxoplasma gondii 250 mU/ml; human, mouse

5×107/mouse

131

Eimeria bovis 0.2 (sporozoites) MOI; bovine

- 132

FUNGI

Aspergillus fumigatus 5 (conidia); human - 132

Candida albicans 0.01 MOI; human - 133

Aspergillus nidulans 0.5 (conidia) MOI; human - 113

VIRUS

Human Immunodeficiency Virus (p24 antigen)

1.0-2.4 ng/mL; human - 134

Influenza A virus H1N1 20 MOI; human 100-500 PFU/mouse 135

Influenza A virus H3N2 2 MOI; mouse 2×105 PFU/mouse

136

HOST FACTORS

GM-CSF + C5a 25 ng/ml GM-CSF + 10

-7 M C5a

- 137

Page 29: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

15

IL-8 2.5–10 ng/ml; human 138

MIP-2

Singlet oxygen 10 μg/ml Photofrin; human - 139

Platelet activating factor (PAF) 10−10

-10−7

M; human 115

Syncytiotrophoblast microparticles (STBM)

150 μg/ml; human 138

Human β-defensin 1 100 ng/ml; human 140

OTHERS

Glucose oxidase 200-1000 mU/mL; human - 115

Calcium ionophore 5 μg/ml; zebrafish

4 μM; human

- 141,142

Phorbol myristate acetate (PMA) 25 nM - 100 nM; human - 100,117

Bacterial component LPS 100 ng/ml; human 5-25 μg/mouse 100,118,121,124

P. aeruginosa Pyocyanin 10-30 μM; human 143

Adapted and modified from Cheng OZ & Palaniyar N120

.

1.3.3 Neutrophil Extracellular Traps in Disease

Neutrophils are key effectors of innate immune and inflammatory responses. They play a key

role in recognition and elimination of pathogens. The resolution of inflammation is required to

limit the destruction caused by neutrophil activation. It is an active process where apoptotic

neutrophils are removed by macrophages and additional leukocyte infiltration is prevented (See

Neutrophil Activation and Resolution 1.3.1). This process is essential for maintaining tissue

homeostasis144

. If impeded, this process can trigger “non resolving inflammation”, a problematic

condition that contributes to pathology. The participation of neutrophil in mediating acute and

chronic inflammation is well documented145

. However, the involvement of NETs in contributing

to inflammatory damage is a relatively new field. This section will highlight the current opinion

of NETs in the context of disease.

Neutrophil extracellular traps are released in response to various activating stimuli and

effectively ensnare and kill bacteria. They are an effective means for antimicrobial clearance as

bacteria that express DNases as virulence factors are more effective at evading host immune

Page 30: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

16

machinery125,127

. This may point to evolutionary pressure to avoid NETs. In addition, restoration

of NADPH oxidase through gene therapy in a patient suffering from CGD cleared persistent

Aspergillus infection and the patients neutrophils demonstrated NET-mediated killing ex vivo113

.

However, NETs do come at a cost. Armed with a hefty arsenal of antimicrobial and

cytotoxic compounds, the increased local concentrations of these compounds acts as a double

edged sword146

. There is no discrimination between host or microbe. The mechanisms for NET

clearance are currently not fully understood, but ineffective clearance can cause deleterious

inflammation on host tissue. NETs have been associated with endothelial and tissue damage as

demonstrated in vitro through propidium iodide staining with human umbilical vein endothelial

cells (HUVEC)118

. The same effect could be observed in vivo in liver microvasculature as NETs

induced a decrease in the number of sinusoids that were perfused and increased serum levels of

alanine aminotransferase (ALT)118

. Furthermore DNA-histone complexes can directly cause

epithelial and endothelial cell death147,148

. In vivo administration of histones resulted in

vacuolated endothelium, intra-alveolar hemmorage, and thrombosis in both micro and macro

vasculature147

. Since their first discovery, NETs have now been implicated in a variety of

inflammatory and autoimmune disease.

1.4 Complement and NETs in Disease

The advent of pharmacological inhibitors and animal models has shed light on our understanding

of the role complement and NETs play in diseases. In this section, we highlight the involvement

of complement and NETs in several inflammatory diseases (Figure 3).

Page 31: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

17

Figure 3. Complement and NETs lead to endothelial cell damage. Failure to regulate the inflammatory response can lead to destruction of host tissues. (A)

Overwhelming endogenous activation, or dysregulation of complement activation through

mutation or auto antibodies can lead to a uncontrolled amplification of complement alternative

pathway leading to tissue damage. (B) The cytotoxic DNA-protein complexes in NETs, which

include neutrophil elastase (NE), myeloperoxidase (MPO), citrullinated histone H3 (CitH3), and

proteinase 3 (PR3), can directly cause tissue damage or lead to the presentation of antigens to

form auto antibodies, anti-citrullinated peptide antibody (ACPA) and anti-neutrophil cytoplasmic

antibody (ANCA).

Page 32: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

18

Lupus Nephritis

Systemic lupus erythematosus (SLE), or lupus, is a autoimmune disorder that effects multiple

organs with a broad range of clinical manifestations149

. Autoantibody initiated activation of

complement is involved in the pathogenesis of tissue damage in SLE. Evidence for this was first

illustrated when administration of anti-C5 monoclonal antibodies ameliorated the development

of glomerulonephritis in the (NZB × NZW)F1 mouse model of SLE52

. Following this, direct

support for the role of complement AP in disease pathogenesis was discovered as CFB-/-

and

CFD-/-

mice are protected from renal disease in the MRL/lpr mouse model for SLE150,151

. The

level of protection was similar to that found in mice treated with C3 inhibitor Crry152

.

Interestingly, elimination of complement CP activation through C4 blockade in models of SLE

led to an increase in an autoimmune response to auto antigens exacerbating tissue damage153

,

thus highlighting the importance of complement AP in contributing to disease pathology.

The formation of NETs, which contain antimicrobial peptides, DNA and histones has

been associated with the presentation of self-antigens leading to the formation of autoantibodies

in SLE. Post-translational modification of histones within NETs drives the process of auto

immunity as serum autoantibodies from SLE patients recognize many post-translationally

modified histones found in NETs154

.

To further exacerbate the issue, serum from SLE patients cannot remove NETs in a

timely manner, thus allowing for presentation of self-antigens. Impaired NET degradation is a

result of DNase I inhibitors, and anti NET auto-antibodies which prevent DNase I from access to

NETs. Prolonged exposure, of affected skin and kidney from lupus patients, to infiltrating

neutrophil extracellular traps can promote damage to host endothelial cells155

. This correlated

with clinical studies as patients who were unable to degrade NETs have significantly higher anti-

dsDNA and anti nucleus antibody titers, a hallmark test for SLE diagnosis, compared to those

who were able to degrade NETs156

. In addition, serum C1q has been shown to be able to bind

NETs. This suggests that in the presence of impaired NET degradation, NETs activate

complement through CP further amplifying the inflammatory response and exacerbating SLE

pathology157

.

Page 33: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

19

Rheumatoid Arthritis

Rheumatoid arthritis is an autoimmune disease characterized by chronic systemic inflammation.

It may affect many tissues and organs, but the principle point of focus is synovial joints158

.

Rheumatoid arthritis has been linked to a single nucleotide polymorphism in the gene for C5

which results in increased concentrations of C5 in serum159

. The implications for AP

involvement in disease progression was illustrated using the K/BxN-derived anti GPI serum

transfer model of Rheumatoid arthritis. In this model, inflammatory joint disease was

ameliorated in CFB-/-

C57BL/6 receiving anti GPI serum but not in C4-/-

mice160

. In the collagen-

induced model of arthritis, CFB-/-

× DBA1/j displayed substantial disease amelioration. These

models again illustrate role complement AP plays disease pathogenisis161

.

In the same K/BxN-murine autoantibody mediated model of arthritis, PAD4 activity is

readily detectable in inflamed synovium of wild type mice. This effect was abrogated when

PAD4 -/-

mice were injected with K/BxN sera to induce disease, suggesting NETs play a

prominent role in disease. Interestingly, PAD4-/-

mice, when compared to PAD4 wt mice, were

not protected from disease pathogenesis. Both mice displayed comparable severity and kinetics

with no statistical significant differences in clinical scores for swelling, joint erosion or

invasion162

. This indicates causes leading to RA disease progression are multi factorial, NETs

may play just one part of many in disease progression.

The presence of auto antibodies mounting immune responses to self antigens is also

prevalent for rheumatoid arthritis patients. Citrullinated proteins are target of anti citrullinated

peptide antibodies (ACPA) in rheumatoid arthritis. Analysis of sera from rheumatoid arthritis

patients revealed that citrullinated H4, from activated neutrophils and NETs, was a target for

ACPA in rheumatoid arthritis163

, suggesting that during NETosis, the presentation of self

antigens can mount for an unwanted immune response.

Asthma

Asthma is a chronic inflammatory disease of the lung airways characterized by recurring

symptoms of airflow obstruction, and bronchospasm. Because complement infiltration out of the

vasculature is poorly understood, asthma is typically associated with hypersensitivity due to IgE

production, cellular immune abnormalities and generation of Th2 cytokines164

. Interestingly, as

Page 34: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

20

evidence for the involvement of C3a anaphyltoxin in asthma pathogenesis, guinea pigs with C3a-

receptor (C3aR) defect demonstrate decreased bronchoconstriction165

. Furthermore in a clinical

study, C3a and C5a levels are elevated in bronchoalveolar lavage fluids from asthma patients166

.

In addition, in mouse models of airway hyper-sensitivity, CFB-/-

mice or wild-type mice treated

with monoclonal antibody to complement factor B demonstrated less inflammation and airway

sensitivity to methacholine when compared to wild type controls. C4-/-

mice however were not

protected against injury167

. Therefore, the complement AP, but not classical or lectin pathway,

plays a role in contributing to asthma disease.

Along with chronic inflammation, asthma is also associated with an influx of leukocytes

into the airways. The presence of neutrophilic asthma (e.g. greater proportion of neutrophils than

other leukocytes) dictates greater disease severity and poor prognosis168,169

. In a recent study, out

of 8 potential biomarkers (IL-8, neutrophils, eosinophils, IL-1Rα, IL-1α, IL-5, IL-6, and

RANTES) only neutrophils and IL-8 could be used to distinguish moderate from mild to severe

asthma in patient bronchoalveolar lavage fluid120

. Moreover, a clinical study has identified the

presence of neutrophil extracellular traps in allergic asthmatic airways. In these patients, both IL-

8, neutrophil count and NETs are shown to be increased in asthmatic airways170

. IL-8 has

previously been shown to be a potent activator of NETs138

. These findings suggest that IL-8,

neutrophil count and NETs may contribute to asthma. At this time, however, the mechanism for

NET formation in the asthmatic airway as well as their contribution to disease severity is not

clearly understood.

Vasculitis

Vasculitis is a group of disorders characterized by inflammation of blood vessels leading to

fibrinoid necrosis of vessel walls. Recent studies have shown that complement AP plays an

important role in anti neutrophil cytoplasmic antibody (ANCA) associated form of vasculitis. In

a mouse model for ANCA-vasculitis, anti MPO-IgG or MPO-reacting splenocytes are obtained

from MPO knockout mice immunized with purified mouse MPO. The anti MPO-IgG or MPO-

reacting splenocytes were then transferred into a recipient mouse171

. In this model of ANCA-

associated vasculitis, complement depletion with cobra venom factor completely blocked the

development of glomerulonephritis and vasculitis induced by splenocyte transfer or MPO IgG

injection. Furthermore, using this model for ANCA-vasculitis, C5-/-

-and CFB-/-

were protected

Page 35: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

21

from disease induction, however C4-/-

mice developed glomerulonephritis and vasculitis similar

to wild-type172

. This rescue effect was not limited to knockout mice models. Mice treated with

anti-C5 antibody 8 h or one day prior to MPO IgG injection showed no signs of disease

development. In addition, antibody administration one day after MPO-IgG injection resulted in

80% reduction of disease pathogenesis173

.

The presentation of ANCA is strongly linked to small vessel vasculitis (SVV). ANCA has an

activating effect on cytokine-primed neutrophils174

. Activation of TNF-α primed neutrophils with

ANCA leads to the formation of NETs175

. In addition, in situ studies, illustrate that NET and

NET proteins are located in close proximity to neutrophil infiltrates in affected glomeruli and

intersitium of SVV patients175

. Furthermore, NETs contain auto antigens proteinase-3 (PR3) and

myeloperoxidase (MPO) which can activate the inflammatory immune response. The persistent

deposition of NETs in inflamed kidneys and circulating MPO-DNA complexes in SVV patients

highlights the involvement of NETs for vasculitis disease pathogenesis.

Sepsis

Severe sepsis can be detrimental, killing approximately 300,000 to 500,000 North Americans a

year176

. In sepsis, infection with microorganisms can trigger acute inflammatory reactions

associated with cytokine storm and influx of other inflammatory mediators. The result can be

drastic producing hypotension, multi-organ failure and even death. Complement activation in

response to infection is important for immune function, however it must be tightly regulated. In

late stages of sepsis, complement C5a can contribute to organ damage through interaction with

other immune cells or induction of coagulation by C5a mediated expression of tissue factor,

which can lead to thrombocytopenia177

. Often the more profound the thrombocytopenia, the

more severe the sepsis and greater the mortality178

.

Investigations of cross-talk between platelets and neutrophils revealed that TLR4

activated platelets can trigger NETosis118

. LPS or plasma from severely septic patients triggered

platelet TLR4 dependent interaction with neutrophils, leading to the production of NETs. Platelet

derived β-defensin 1, was further demonstrated to be able to activate neutrophils to form

NETs140

. In this setting, the formation of NETs allowed for efficient aid in bacterial trapping

during septic conditions, however, NETs also contributed to tissue damage. Platelet-induced

NETs were associated with hepatotoxicity due to tissue damage118

. This could account for some

Page 36: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

22

of the mechanisms contributing to organ failure during sepsis. Furthermore, NETs have also

been demonstrated to display a broad range of procoagulant properties. NETs induce the

formation of a red blood cell-rich thrombi by serving as a scaffold for platelet/RBC adhesion and

aggregation. NETs also serve as a platform binding plasma proteins to promote thrombus

stability179

. In addition, NETs also directly promote thrombus formation as presentation of

nuclear histones promotes thrombin generation through platelet-dependant mechanisms

involving TLR2 and TLR4180

.

1.5 Complement and Neutrophils

The activation of complement leads to the generation of anaphylatoxins C3a and C5a. These

anaphylatoxins initiate a strong inflammatory response and result in leukocyte influx. Generation

of C3a and C5a gradients guide neutrophils to sites of inflammation through chemotaxis181

. In

addition, these anaphylatoxins can serve as ligands binding neutrophil receptors to trigger

neutrophil activation. The activation of neutrophils with C5a usually does not induce NETosis,

however, neutrophils primed with host-derived cytokine, granulocyte-macrophage colony

stimulating factor (GM-CSF), are able to release NETs of mitochondrial origin when activated

with C5a137

. In experimental models, acute inflammatory states in C5-deficient mice result in

reduced neutrophil influx, often associated with attenuated tissue damage. This underlies the

importance of C5aR in mediating the inflammatory response182

.

Neutrophils express both C3a receptor (C3aR) and C5a receptor (C5aR) on their plasma

membranes183,184

. C3aR and C5aR are both classical G protein-coupled receptors. Binding of

ligands to these receptors activates G-protein coupled signaling transduction pathways which

lead to neutrophil chemotaxis and activation. Response to complement anaphylatoxins must be

tightly regulated in situations with sustained complement activation to avoid deleterious effects.

Modulation of neutrophil response to C3a and C5a is achieved through receptor

internalization185

. Binding of C3a and C5a to their respective receptors leads to a ligand-

mediated internalization of the receptor that is modulated by phosphorylation of the C-terminal

domains185,186

. C3a activates the internalization of C3aR and C5a activates the internalization of

C5aR, there is no cross-internalization of receptors185

.

Upon activation, neutrophil granules are mobilized to the surface releasing their cytotoxic

contents at sites of inflammation. The distinction between the different classes of neutrophil

Page 37: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

23

granules is not only important in characterizing their constituents, it also reflects on their ability

to mobilize. Granules formed latest having the highest propensity for degranulation, thus

illustrating the neutrophils ability to modulate inflammatory responses187

. The neutrophil

secretory vesicles are mobilized completely by many activating stimuli. They are stores of

adhesion molecules, which upon activation, can mobilize to the surface to initiate leukocyte

adhesion, rolling or transmigration188

. Secretory vesicles also serve as an intracellular reservoir

for complement receptors. There is evidence that secretory vesicles contain C1q Receptor

(C1qR), complement receptor 1 (CR1), complement receptor 3 (CR3), and complement receptor

4 (CR4)189

.

Mobilization of complement receptors to neutrophil surfaces allows for phagocytosis of

complement opsonized particles (e.g. C1q, C3b, or C4b) through IgG independent mechanisms,

thus linking neutrophils to innate immunity190

. Moblization of C1qR mediates enhancement of

C1q opsonized particles for phagocytosis191

. Mobilization of CR1 mediates phagocytosis of

complement opsonized particles. It is also responsible for regulating complement amplification.

In addition, uptake of adenovirus is attributed to CR1, not CR3 or CR4192

. CR3 mobilization

allows for adhesive interaction between the neutrophil complement receptor and its endothelial

cell ligand ICAM-1. This allows for neutrophil adhesion and further activation98

. CR3 is also

responsible for phagocytosis of complement opsonized particles in a urokinase receptor

dependent manner193

. In addition, CR3, demonstrates unique interactions with CFH and CFH

related protein 1 (CFHR1). CFH and CFHR1 binding to neutrophils via CR3 mediates

attachment to Candida albicans and confers antimicrobial resistance194

. CR4 facilitates the

phagocytosis of complement opsonized components190

.

In addition to complement receptors, neutrophils also express complement regulatory

proteins. Neutrophils express DAF, CD59, and MCP on their plasma membrane surfaces.

Expression of complement regulatory proteins can be altered in response to injury195

.

1.6 Neutrophil Complement Cross-talk

Complement activation leads to the formation of powerful anaphylatoxins which can guide

neutrophils to sites of inflammation through chemotaxis. There is, however, a growing body of

evidence to suggest that neutrophils can also initiate complement amplification. Addition of

neutrophil hydrogen peroxide to serum leads to a diminution in serum hemolytic activity as well

Page 38: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

24

as reduction in complement C3 and C5. In addition, complement activation products, C3a and

C5a, were produced in serum treated with hydrogen peroxide. This effect could be abolished

with EDTA but not EGTA indicating that neutrophil oxygen radicals can amplify the

complement alternative pathway196

.

In a separate study, purified MPO was able to convert C5 into an activated form which

led to the formation of C5b-9. The mechanism for activation of C5 was dependent on MPO

generation of HOCl from hydrogen peroxidase. This leads to the oxidation, thus activation of C5.

Supernatant from activated neutrophils was also able to activate complement through MPO-

generated hyplocholrite dependent mechanisms197

. Neutrophil elastase was also demonstrated to

be able to cleave C5 to form C5b-941

.

Furthermore, neutrophils are a potent source of properdin, the only positive regulator of

complement AP. CFP is stored in neutrophil secondary granules and is secreted upon

activation198

. Activation of neutrophils can increase local concentrations of CFP, thus amplifying

complement AP. Infiltrating neutophils can often trigger unnecessary inflammatory responses

through propagation of complement AP83

. In addition secreted properdin can bind to activated

neutrophils. This process initiates the formation of complement convertases which ultimately

leads to the formation of C5b-9 on neutrophil surfaces. The result is the production of

anaphylatoxins and release of neutrophil cytokines which further directs infiltration of additional

neutrophils. Thus activated neutrophils amplify complement AP in a positive feedback loop199

.

Page 39: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

25

1.7 Rationale

The persistence of both neutrophil NETs and complement amplification in many deleterious

autoimmune or inflammatory diseases suggests that their interactions may exacerbate the

inflammatory immune response. Evidence shows that there is extensive cross-talk between

neutrophil activation and complement activation. However, not much is known within the

context of NETs (e.g. interaction of complement proteins with NETs).

It is interesting to note that DNA exposed on apoptotic and necrotic cells is able to bind

C1q and MBL. Binding of MBL to apoptotic cells enhances phagocytosis by macrophages but

does not activate complement200

. Binding of C1q triggers basal levels of complement activation,

however, the process is often simultaneously inhibited through C4 binding protein (C4BP) and

CFH201

. The binding of these inhibitory proteins not only limits complement activation but

inhibits further release of DNA202

. These inhibitory mechanisms maybe overwhelmed in the

presence of NETS.

NETs have been shown to amplify complement, however the mechanisms for activation

remain elusive. NETs are able to bind C1q which suggest that they may activate complement

through CP157

. NETs, also contain MPO which has been shown to activate complement AP197

. In

addition, neutrophil elastase, a known NET protein, is able to cleave C5 to activate

complement41

. All of these remain viable in explaining the mechanism for neutrophil NET

complement amplification.

In addition, neutrophil CFP has well documented pattern recognition molecule properties.

CFP interacts with a variety of polyanionic macromolecules including DNA expressed on

necrotic or apoptotic cells, however, the interaction with extracellular DNA on NETs has not yet

been investigated. Properdin opsonized targets have potential to amplify complement. In essence,

a complement activating surface is any surface without adequate regulatory protein function to

control the complement AP, or unable to control AP via CFH203

.

Taken together, NET and complement interactions could play a significant role for

antimicrobial activity or in the pathogenesis of many inflammatory and auto immune diseases

(Figure 4). To date, there has been no evidence detailing the mechanisms in which NETs amplify

the complement.

Page 40: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

26

Figure 4. Complement interaction with NETs.

Neutrophils are key mediators of innate immune and inflammatory responses. Neutrophil influx

to sites of inflammation can amplify complement activation through release of properdin (CFP).

Neutrophil elastase (NE) and myeloperodiase (MPO) can also contribute to complement

activation. Neutrophils also form NETs to efficiently capture invading microbes, however, the

mechanisms for microbial killing are not fully understood. We propose that neutrophil

extracellular traps can activate complement to enhance microbial clearance. If unregulated, this

process can lead to endothelial cell damage. The dashed line in the center divides the paradoxical

roles of how complement and neutrophil interactions may lead to antimicrobial clearance or

endothelial cell damage.

Page 41: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

27

1.8 Hypothesis

Complement factors present in neutrophils participate in antimicrobial function by mounting a

targeted response to infectious agents via NETs by localized complement alternative pathway

activation.

1.9 Aims

Aim 1. To characterize the interaction between the complement system with neutrophils and

neutrophil extracellular traps.

Aim 2. To determine the significance of complement-NET interactions in antimicrobial function.

Page 42: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

28

2 Methods

2.1 Reagents

All buffer salts and reagents were obtained from Sigma Aldrich (St. Louis, Mo, USA) unless

otherwise stated.

2.2 Research Ethics Board Approval

Informed and written consent was obtained from the donors and the protocol was approved by

the ethics committee at the Hospital for Sick Children.

2.3 Neutrophil Isolation

Human peripheral whole blood was collected from healthy individuals and aliquoted into BD

vacutainer containing EDTA (1.8 mg/ml). 20 ml of whole blood was layered on top of 20 ml of

Polymorphprep™ (Axis-shield, Oslo, Norway) and separation was achieved by centrifugation at

500 × g for 35 minutes, allowing the rotor to decelerate without brake. The lower band

containing polymorphonuclear cells was harvested and resuspended in an equal volume of a half

strength Hepes-buffered saline (0.425% (w/v) NaCl, 5 mM Hepes-NaOH, pH 7.4) returning the

polymorphonuclear cell suspension to isoosmotic condition. Neutrophils were harvested by

centrifugation at 400 × g for 10 minutes. Removal of residual erythrocyte contamination was

achieved through hypotonic lysis. Polymorphonuclear cell pellets containing erythrocytes were

resuspended in a hypotonic solution (0.2% (w/v) NaCl) for 30 seconds followed by addition of

an equal volume of hypertonic solution (1.6% (w/v), 20 mM Hepes-NaOH, pH 7.4). Following

hypotonic lysis of erythrocytes, the polymorphonuclear cells are washed twice in Hepes-buffered

saline (0.85% (w/v) NaCl, 10 mM Hepes-NaOH, pH 7.4). Cells were harvested and resuspended

in the desired media. Cell counting was performed with a hemocytometer and cells were

resuspended to the desired concentration.

2.4 Preparation of Neutrophil Lysates

Neutrophils were harvested via centrifugation at 1000 × g for 10 minutes. The neutrophil cell

pellet was resuspended in a neutrophil lysis buffer (1% (v/v) Triton X-100, 50 mM Tris, pH 7.4,

10 mM KCl) containing 2 × cOmplete, mini protease inhibitor cocktail (Roche Diagnostics,

Laval, QC, Canada) supplemented with 0.5 mM EDTA, 25 μM leupeptin, 25 μM pepstatin, 25

Page 43: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

29

μM aprotinin, 1 mM levamisole, 1 mM Na3VO4, 25 mM NaF, 1 mM PMSF. The sample was

sonicated 3 times for 10 seconds (VWR Sonics model 50D) and incubated for 15 minutes at 4°C.

Neutrophil lysates were centrifuged at 25000 × g for 30 minutes at 4°C and stored at -80°C for

future analysis. Neutrophils were resuspended to a final concentration of 2 × 107 cells/ml.

2.5 Bacterial Cultures and Strains

Bacteria were cultured from glycerol stocks kept at -80°C or from single colonies selected from

LB agar plates. P. aeruginosa mPAO1 (a gift from the lab of Dr. Neil Sweezey, The Hospital for

Sick Children), P. aeruginosa PAKwt (a gift from the lab of Dr. Marina Ulanova, Northern

Ontario School of Medicine), S.aureus RN6390 (a gift from the lab of Dr. John Brumell, The

Hospital for Sick Children), B. subtilis, and E. coli Y1088 (Bacteria from Dr. Nades Palaniyar,

The Hospital for Sick Children) were cultured in LB agar broth. P. aeruginosa PAKgfp (a gift

from the lab of Dr. Marina Ulanova, Northern Ontario School of Medicine) was cultured in LB

agar broth with 30 μg/ml gentamicin (Wisent Bioproducts, Montreal, QC, Canada).

2.5.1 Bacterial Plating

Agar plates were prepared with autoclaved LB-agar. Bacterial cultures were streaked onto agar

plates and incubated overnight at 37°C. PAKgfp was maintained on LB-agar with 30 μg/ml

gentamicin. Agar plates were wrapped in parafilm and stored at 4°C for up to two weeks.

2.5.2 Bacterial Growth

Bacteria cultures were selected from single colonies on LB-agar and grown overnight in sterile

LB-broth. PAKgfp was maintained in LB-broth with 30 μg/ml gentamicin. Prior to neutrophil

actvation, bacterial cultures were diluted by a factor of ten and sub-cultured for 3 hours. Cells

were harvested by centrifugation at 5000 × g for 5 minutes at 4°C and washed three times in 5 ml

of phosphate-buffered saline (PBS, pH 7.4). After the final resuspension, turbidity of the culture

was taken at OD600 and the concentration of the bacteria was approximated using the formula:

Bacteria was resuspended to the appropriate concentration in PBS and used for neutrophil

activation studies at a multiplicity of infection (MOI) of 10 and MOI of 100.

Page 44: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

30

2.6 Neutrophil Activation

2.6.1 Neutrophil Oxidative Burst

Purified neutrophils as described in the section Neutrophil Isolation (2.3) were resuspended at 1

× 106

cells/ml in RPMI 1640 (Wisent Bioproducts, Montreal, QC, Canada) supplemented with 10

mM Hepes, pH 7.4. Cells were activated with C5a (1-2 μM), fMLP (1-2 μM), and Phorbol 12-

myristate 13-acetate (PMA, 20 nM) for 30 minutes. Dihydrorhodamine 123 (10 μM) was used

for detection of reactive oxygen species.

Cells were analyzed by flow cytometry using a Gallios Flow Cytometer (Beckman

Coulter, Mississauga, ON, Canada). Cells were gated through forward scatter and side scatter to

determine the neutrophil population. The gated population was further stained for with Hoescht

(1 μg/ml) using filter channel: FL9 (405/450 BP 40) to select for nucleated populations. Reactive

oxygen species was detected using filter channel FL1 (488/429 BP 28.25).

2.6.2 Neutrophil Activation and Secretion

Purified neutrophils as described in the section Neutrophil Isolation (2.3) were resuspended in

RPMI 1640 media (Wisent Bioproducts, Montreal, QC, Canada) supplemented with 10 mM

Hepes, pH 7.4 to a final concentration of 2 × 107 cells/mL. Neutrophils were aliquoted into

Eppendorf tubes and activated with C5a (1 μM), fMLP (1 μM) and PMA (20 nM). Cells were

cultured in a tissue culture incubator (37°C, 5% CO2) for 30 minutes with slight agitation every

10 minutes to prevent cell clumping. Neutrophil stimulation was terminated by removing

neutrophils from tissue culture and incubating at 4°C for 5 minutes. Neutrophils were harvested

via centrifugation at 1000 × g for 10 minutes. The supernatant was carefully collected and

centrifuged at 25000 × g for 10 minutes at 4°C and immediately placed in 2 × neutrophil laemmli

sample buffer. The supernatant was heated at 95°C for 5 minutes and stored at -80°C for future

analysis. Neutrophil lysates were prepared from the remaining cell pellet as described in the

section Preparation of Neutrophil Lysates (2.4)

2.6.3 Differential Cell Counts for Nuclear Morphology

Neutrophils as described in section Seeding Neutrophils on Coated Cover Slips (2.8.3) were

activated with 20 nM PMA and fixed at the following time points (t=0, 30, 60, 120, 180, 240

min) with 4% (w/v) paraformaldehyde (Electron Microscopy Sciences, Fort Washington, PA,

Page 45: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

31

USA). Cells were stained with DAPI (1 μg/ml) and cover slips were mounted with Dako

Fluorescence Mounting Media (Dako Canada, Burlington, ON, Canada). Images were taken

using a Nikon Eclipse Ti-U Microscope, equipped with QICAM 12 bit Mono Fast 1394, model:

QIC-F-M-12C camera (Qimaging, Surry, BC, Canada) using a 40×/0.60 air immersion objective

operated by NIS-Elements software (Nikon, Mellville, NY, USA). The nuclear morphologies

were categorized as lobular, delobular, decondensed, NETs, and undefined. Differential cell

counts for nuclear morphology was assessed by counting 118 to 220 cells from 5 different fields

of view at various time points. Nuclear morphology types are presented as a percent total

averaged from three independent experiments.

2.6.4 SYTOX Green Plate Reader Assay

Neutrophils (3 × 104 cells) were seeded onto 96 well plates. For inhibition of NETs, neutrophils

were preloaded with 2 μM of NADPH oxidase inhibitor, diphenyleneiodonium (DPI) for one

hour before activation. Neutrophils were activated with PMA (20 nM), C5a (1 μM), and fMLP (1

μM). To study NETs in response to sepsis, Pseudomonas aeruginosa (mPAO1, PAKwt,

PAKgfp), Staphylococcus aureus (RN6390), Escherichia coli (Y1088), and Bascillus subtilis

were used to activate neutrophils at MOIs of 10 and 100.

Cell impermeable DNA dye, SYTOX Green (5 μM), was added to neutrophil suspensions

to detect the presence of extracellular DNA. When neutrophils undergo NETosis, the release of

NETs can be detected by SYTOX green fluorescence intensity. Fluorescence intensity was

measured using the POLARstar Omega microplate reader (BMG Labtech, Ortenberg, Germany)

with excitation/emission (485/520). NET formation was normalized to total DNA in resting

neutrophils permeabilized with 0.5% (w/v) Triton X-100.

2.7 Western Blot

BCA protein determination assay was used prior to preparation of samples for western blot.

Neutrophil lysates were prepared in 2 × neutrophil laemmli buffer (125 mM Tris-HCl, pH 6.8,

6% (w/v) SDS, 8% (v/v) β-mercaptoethanol, 18% (v/v) glycerol, 10 mM EDTA, 25 μM

leupeptin, 25 μM pepstatin, 25 μM aprotinin, 10 mM NaF, 5 mM Na3VO4, 1mM levamisole,

0.05% (w/v) bromphenol blue) and heated at 95°C for 5 minutes with shaking. For non reducing

samples, neutrophil lysates were prepared in 2 × neutrophil laemmli sample buffer in the absence

Page 46: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

32

of β-mercaptoethanol. 50 μg of protein from neutrophil lysates were loaded and separated by

10% SDS-polyacrylamide gel electrophoresis running at 200 V for 45 minutes. Protein gels were

transferred onto nitrocellulose membrane for 70 minutes at 100 V. 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. For identification of complement proteins, membranes were

incubated with goat polyclonal antibody to complement proteins (1:1000 dilution; Complement

Technology, Tyler, TX, USA) in 5% (w/v) skim-milk in TBST overnight at 4°C with rocking.

For loading controls, membranes were incubated with mouse monoclonal antibody to β-actin (β-

actin, BA3R, 1:10000 dilution; Thermo Fisher Scientific, Rockford, IL, 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 5 times with TBST

for 5 minutes. Proteins were detected using Western Lighting™ Plus-ECL, Enhanced

(PerkinElmer, Waltham, MA, USA) and developed on radiographic film on a Kodak X-Omat

2000a processor.

2.7.1 Determining Protein Concentration

A bicinchoninic acid (BCA) assay was performed on neutrophil lysates to determine total protein

concentration. BCA protein assay kit was used according to manufacturer protocol without

deviation (Thermo Fisher Scientific, Rockford, IL, USA). 50 μg of protein was loaded onto SDS-

PAGE for western blot.

2.8 Imaging and Microscopy

2.8.1 Acid Washed Cover Slips

Coverslips were acid washed prior to use for microscopy to remove manufacturing residue, oils

and other contaminants such as lipopolysacchardie (LPS) to prevent non specific activation of

neutrophils. 12-mm round glass coverslips (Fisher Scientific) were submerged in nitric acid for

48 hour. Coverslips were washed with deionized tissue-culture-grade water for 15-20 exchanges

or until wash exchanges reach neutral pH, followed by two exchanges of 95% (v/v) ethanol to

remove residual water. Cover slips were stored in 70% (v/v) ethanol for future use.

Page 47: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

33

2.8.2 Preparation of Cover Slips for Microscopy

Acid washed cover slips were removed from ethanol storage and excess ethanol was drained

using Kimwipes. Cover slips were flame dried and placed into a 6-well tissue culture plate.

Three cover slips were placed in each well. Cover slips were coated with 75 μl of 0.001% Poly-

L-Lysine for 60 minutes at room temperature. Cover slips were washed three times with PBS (~3

mL). Poly-L-lysine coated cover slips were used in all experiments prepared for analysis with

micrsocopy.

2.8.3 Seeding Neutrophils on Coated Cover Slips

Purified neutrophils as described in Neutrophil Isolation (2.3) were resuspended in RPMI 1640

media (Wisent Bioproducts, Montreal, QC, Canada) supplemented with 10 mM Hepes, pH 7.4,

to a concentration of 1 × 106 cells/ml. 1 ml of the neutrophil suspension was placed in tissue

culture dish containing the coated cover slips (2.8.3). Neutrophils adhere to cover slips after 1

hour incubation (37°C, 5% CO2).

2.9 Identifying Complement Proteins in Neutrophils

Neutrophils were prepared for microscopy as described in Seeding Neutrophils on Coated Cover

Slips (2.8.3) and activated with PMA (20 nM) for 30 minutes (37°C, 5% CO2). No exogenous

plasma or serum was added to the culturing media. Samples were fixed with 4% (w/v)

paraformaldehyde (Electron Microscopy Sciences, Fort Washington, PA) in PBS. Samples were

stained with 5 μg/mL Wheat Germ Agglutinin, Alexa Fluor® 555 conjugate (Invitrogen, Eugene,

OR, USA) for 10 min and washed in PBS. Samples were permeabilized with 0.2% (v/v) Triton

X-100 for 20 minutes, and washed in PBS. Samples were blocked in 3% (v/v) gelatin from cold

water fish skin (v/v), and stained with anti complement antibodies: rabbit polyclonal antibody to

properdin (SC-68366, 1:50 dilution; Santa Cruz Biotechnology, Dallas, TX, USA), rabbit

polyclonal antibody to CFB (SC-67151, 1:50 dilution; Santa Cruz Biotechnology, Dallas, TX,

USA), rabbit polyclonal antibody to complement C3 (ab97462, 1:100 dilution; Abcam,

Cambridge, MA, USA), and goat polyclonal antibody to CFH (AF4779, 1:50; R&D Systems,

Minneapolis, MN, USA) for 1h. Samples were washed in PBS and incubated with respective

secondary donkey-anti primary antibodies with Alexa Fluor® 488 conjugate (Invitrogen,

Eugene, OR, USA). Cell nuclei were stained by DAPI (1 μg/ml). Samples were washed in PBS

Page 48: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

34

and cover slips were mounted with Dako Fluorescence Mounting Media (Dako Canada,

Burlington, ON, Canada) for analysis with spinning disc confocal microscopy.

2.10 Detection of Complement Proteins in NETs

Neutrophils were prepared for microscopy as described in Seeding Neutrophils on Coated Cover

Slips (2.8.3) and activated with PMA (20 nM) or P. aeruginosa PAKgfp (MOI of 10) for 240

minutes (37°C, 5% CO2). No exogenous plasma or serum is added to the culturing media. The

samples were fixed with 4% (w/v) paraformaldehyde (Electron Microscopy Sciences, Fort

Washington, PA) in PBS. Samples were blocked with 3% (v/v) gelatin from cold water fish skin

for 1h followed by incubation with anti-complement antibodies: rabbit polyclonal antibody to

properdin (SC-68366, 1:50 dilution; Santa Cruz Biotechnology, Dallas, TX, USA), rabbit

polyclonal antibody to CFB (SC-67151, 1:50 dilution; Santa Cruz Biotechnology, Dallas, TX,

USA), rabbit polyclonal antibody to complement C3 (ab97462, 1:100 dilution; Abcam,

Cambridge, MA, USA), and goat polyclonal antibody to CFH (AF4779, 1:50; R&D Systems,

Minneapolis, MN, USA) for 1h. Neutrophils activated with PMA were co-stained with mouse

monoclonal antibody to neutrophil myeloperoxidase (ab25989, 1:500 dilution; Abcam,

Cambridge, MA, USA). Samples were washed in PBS and incubated with secondary donkey

anti-primary antibodies conjugated with Alexa Fluor ®555 (Invitrogen, Eugene, OR, USA).

PAKgfp signal was enhanced with mouse monoclonal antibody to GFP (A11120, 1:100 dilution;

Invitrogen, Eugene, OR, USA) primary for 1h, washed in PBS, followed by incubation with

donkey anti-mouse conjugated with Alexa Fluor ® 488 (Invitrogen, Eugene, OR, USA). NETs

were detected by DAPI (1 μg/ml) staining. Samples were washed in PBS and cover slips were

mounted with Dako Fluorescence Mounting Media (Dako Canada, Burlington, ON, Canada) for

analysis with spinning disc confocal microscopy.

2.11 Complement Competent Plasma

Human peripheral whole blood was collected in 10 μg/ml Refludan® (Bayer Healthcare, Wayne,

NJ, USA) and centrifuged at 1500 × g for 15 minutes. The platelet poor plasma (PPP) was

collected and sterile filtered through 0.2 μm syringe filters. PPP samples were stored at -80°C for

future use. Refludan ® is the pharmacological trade name for lepuridin (recombinant hirudin

derived from yeast cells). It prevents coagulation through inhibition of thrombin. It is

Page 49: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

35

advantageous when collecting complement competent plasma as it does not chelate Ca2+

or

Mg2+

, thus allowing for complement activity.

2.12 Preparation of Rabbit Erythrocytes

Rabbit erythrocytes (Complement Technologies, Tyler, TX, USA) were aliquoted into

Eppendorf tubes and harvested through centrifugation at 6000 × g for 60 seconds. Rabbit

erythrocytes were washed through continuous cycles of centrifugation and resuspension in

complement alternative pathway buffer (AP buffer), containing 20 mM Hepes, pH 7.4, 144 mM

NaCl, 7 mM MgCl2, and 10 mM EGTA, until supernatant is clear. Cells are resuspended to a

final concentration of 5 × 108 erythocytes/ml.

2.13 Plasma Complement Activity Assay

To inhibit complement AP, mouse monoclonal antibody to properdin (Anti Factor P#1, A233;

Quidel Corporation, San Diego, CA, USA) was serially diluted in 40% (v/v) Refludan® (Bayer

Healthcare, Wayne, NJ, USA) fresh frozen PPP/AP buffer. 50 μl of the 40% (v/v) plasma/AP

buffer, containing inhibitory antibody, was added to 50 μl of freshly prepared rabbit erythrocytes

(5 × 108 erythrocytes/ml) in AP buffer to give a final concentration of 20% (v/v) plasma/AP

buffer. Plasma and rabbit erythrocytes were incubated for 15 minutes at 37°C. Residual

erythrocytes were removed through centrifugation at 6000 × g for 60 seconds. 100 ul of

supernatant from each reaction was added to a 96 well tissue culture dish. Free hemoglobin was

measured in supernatants with end point absorption at 405 nm on a POLARstar Omega

microplate reader (BMG Labtech, Ortenberg, Germany). Samples were normalized to total

hemolysis of 2.5 × 107 erythocytes (50 μl) of rabbit erythrocytes in 100 μl of distilled water.

2.14 Complement Activation and C5b-9 Formation on NETs

Neutrophils were prepared for microscopy as described in Seeding Neutrophils on Coated Cover

Slips (2.8.3) with slight deviation. A single cover slip was placed in a 12 well tissue culture plate

and 500 μl of the neutrophil suspension (1 × 106 neutrophils/ml) in RPMI 1640 (Wisent

Bioproducts, Montreal, QC, Canada) supplemented with 10 mM Hepes, pH 7.4 was placed in

each well. Neutrophil samples were preloaded with DNase I (50 μg/ml) to digest all extracellular

DNA including NETs and activated with PMA (20 nM) or P. aeruginosa PAKgfp (MOI of 10)

for 240 minutes (37°C, 5% CO2).

Page 50: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

36

After 240 minutes of activation, tissue culture plates were centrifuged at 200 × g for 5

minutes 4°C. The media was carefully removed and 500 μl of 20% (v/v) Refludan® (Bayer

Healthcare, Wayne, NJ, USA) fresh frozen PPP prepared in RPMI 1640 media or AP buffer was

added to the 12 well tissue culture plate. For experiments with AP buffer, a buffer exchange was

performed with 3 washes with PBS, followed by one wash with AP buffer, to remove

phosphates, prior to the addition of 20% (v/v) Refludan® (Bayer Healthcare, Wayne, NJ, USA)

fresh frozen PPP in AP buffer. Samples were incubated for 15 minutes (37°C, 5% CO2) and

fixed with 4% (w/v) paraformaldehyde (Electron Micrscopy Sciences, Fort Washington, PA) in

PBS.

Samples were blocked with 3% (v/v) gelatin from cold water fish skin for 1h and

incubated with mouse monoclonal antibody to C5b-9 (DIA 011-01, 1:200 dilution; Antibody

Shop, Gentofte, Denmark) for 1h. Samples were washed in PBS and incubated with donkey anti-

mouse secondary antibody conjugated with Alexa Fluor ® 555 (Invitrogen, Eugene, OR, USA).

PAKgfp was enhanced with Alexa Fluor ® 488 conjugated rabbit polyclonal antibody to GFP

(A21311, 1:400 dilution; Invitrogen, Eugene, OR, USA). NETs were detected by DAPI (1

μg/ml) staining. Samples were washed in PBS and cover slips were mounted with Dako

Fluorescence Mounting Media (Dako Canada, Burlington, ON, Canada) for analysis with

spinning disc confocal microscopy.

2.15 Inhibiting Properdin Activity in NETs

Purified neutrophils were prepared for addition of 20% (v/v) Refludan® (Bayer Healthcare,

Wayne, NJ, USA) fresh frozen PPP in RPMI 1640 media or AP buffer as described in

Complement Activation and C5b-9 Formation on NETs (2.14). For inhibition of properdin

activity, mouse monoclonal antibody to properdin (Anti Factor P#1, A233; Quidel Corporation,

San Diego, CA, USA) was added to 20% plasma at a concentration previously determined using

the procedure outlined in Plasma Complement Activity Assay (2.13). Samples were incubated

for 15 minutes (37°C, 5% CO2) and fixed with 4% (w/v) paraformaldehyde (Electron Micrscopy

Sciences, Fort Washington, PA) in PBS.

Samples were blocked with 3% (v/v) gelatin from cold water fish skin for 1h and

incubated with rabbit polyclonal antibody to C5b-9 (ab55811, 1:1000 dilution; Abcam,

Cambridge, MA, USA). Samples were washed in PBS and incubated with donkey anti-rabbit

Page 51: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

37

secondary antibody conjugated with Alexa Fluor ® 555 (Invitrogen, Eugene, OR, USA). NETs

were detected by DAPI (1 μg/ml) staining. Samples were washed in PBS and cover slips were

mounted with Dako Fluorescence Mounting Media (Dako Canada, Burlington, ON, Canada) for

analysis with spinning disc confocal microscopy.

2.16 Spinning-Disc Confocal Microscopy

Images were taken on an Olympus IX81 inverted fluorescence microscope using a 60×/1.35 oil

immersion objective equipped with a Hamamatsu C9100-13 back-thinned EM-CCD camera and

Yokogawa CSU X1 spinning disc confocal scan head (with Spectral Aurora Borealis upgrade).

The unit is equipped with 4 separate diode-pumped solid state laser lines (Spectral Applied

Research, 405 nm, 491 nm, 561 nm, 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 disc confocal microscope was

deconvolved by iterative restoration using Volocity Software (PerkinElmer, Waltham, MA,

USA) with confidence limit set to 95% and iteration limit set to 20.

2.17 Colocalization Analysis

Colocalization analysis was performed using Volocity Software (PerkinElmer, Waltham, MA,

USA). Thresholds were selected to the background and thresholded Pearson’s correlation was

recorded as measure of colocalization.

2.18 Statistical Analysis

Student t-test or one-way/two-way ANOVA with Tukey’s multiple comparison test was used for

statistical comparison as needed. A p value was set at 0.05, 0.01 or 0.001 for statistical

significance. All statistical analysis was performed using GraphPad Prism(GraphPad Software,

La Jolla, CA, USA) statistical analysis software (Version 6.0).

Page 52: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

38

3 Results – Aim 1

To characterize the interaction between the complement system with neutrophils and neutrophil

extracellular traps.

3.1 Neutrophils Contain Complement Factors

The first aim of our study was to characterize the interaction between complement proteins and

neutrophils. Infiltrating neutrophils are often associated with amplification of complement

response leading to inflammatory damage83

. However, the mechanisms in which neutrophils

amplify complement activation are not fully understood. Existing literature suggests that

neutrophils are key mediators of complement AP because they are a potent source of

properdin198

. In addition, early reports have shown that neutrophils may also contain complement

factor B, complement components C3, C6 and C7204

, however these early reports have not

gathered much attention. There is currently a large gap in knowledge surrounding complement

proteins and neutrophils.

To consolidate the data from existing literature, we performed a series of western blots to

identify complement proteins that are contained in human neutrophils. Human neutrophils were

isolated from healthy donors using polymorph prep. Neutrophil lysates were prepared at a final

concentration of 2 × 107 neutrophils/ml. Total protein concentration was determined using a

BCA assay, and 50 μg of protein was loaded onto SDS-PAGE for western blot.

Consistent with the literature, western blot analysis revealed that neutrophils express

properdin (CFP) (Figure 5A), complement component C3 (C3) (Figure 5B), and complement

factor B (CFB) (Figure 5C). In addition, we were able to demonstrate that neutrophils contain

complement factor H (CFH) (Figure 5D) the major soluble regulator of complement AP. To our

knowledge, this is the first report of neutrophils as a source of CFH. All complement antibodies

were tested on diluted human plasma which is an abundant source for complement proteins. The

antibodies for CFP, C3, CFB and CFH all yielded a strong specific band in human plasma

diluted 1:50 (plasma:PBS) after 5 seconds of exposure on radiographic film. This suggests

neutrophils contain a very high concentration of CFP, as we were able to observe a distinct band

on radiographic film after 5 seconds of exposure. The concentrations for C3, CFB and CFH were

much lower as distinct bands only visible on radiographic film after 5 minutes of exposure. In

Page 53: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

39

addition, using western blot analysis, we screened for complement components C1q, C2, C4, C5,

C6, C7, C8, C9, CFD, and CFI, however we were unable to detect the presence of these proteins

(Data not shown).

The presence of CFP, C3, CFB and CFH in resting neutrophils is consistent with the observation

that neutrophils are key mediators of complement alternative pathway. CFP, C3, and CFB are all

major components required to form the complement AP C3 convertase. In the presence of

circulating CFD (1-9 μg/ml), neutrophils express all the necessary constituents to activate

complement AP205

. The presence of CFH in neutrophils suggests that neutrophils may undergo

some form of self regulation of complement activity. This further strengthens the notion that

neutrophils are key mediators of complement AP.

Page 54: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

40

Figure 5. Neutrophils contain proteins from complement alternative pathway.

Representative western blot for detection of complement proteins contained in resting

neutrophils. 50 μg of protein from neutrophil lysates was used for detection of complement

protein. (A) CFP, 10% (w/v) non reducing SDS-PAGE, 5 sec exposure on radiographic film. (B)

C3, 10% reducing SDS-PAGE, 5 min exposure on radiographic film. (C) CFB, 10% reducing

SDS-PAGE, 5 min exposure on radiographic film. (D) CFH, 10% (w/v) non-reducing SDS-

PAGE, 5 min exposure on radiographic film.

Page 55: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

41

3.2 Investigating Neutrophil Activation

Neutrophils are dynamic cells which traverse through the blood stream continuously and

randomly probing endothelium of vessel walls. Upon activation, neutrophils rapidly degranulate

to modify expression of adhesion molecules on the plasma membrane and secrete proteins into

the extracellular environment. To further characterize the interaction between complement and

neutrophils, we examined the localization and protein secretion for complement proteins we

previously identified (Section 3.1) in response to various activating stimuli. From a panel of

activating agonists, we selected two potent mediators of the inflammatory response. We

activated neutrophils with complement anaphylatoxin C5a, and formylated peptide fMLP. In

addition, we also activated neutrophils with PMA, a potent protein kinase-C (PKC) activator.

3.2.1 Neutrophil Activation and Oxidative Burst

Neutrophil activation leads to an NADPH-mediated formation of reactive oxygen species (ROS)

known as an oxidative burst. This process is essential for immune function. We investigated the

effect of C5a, fMLP and PMA on neutrophil ROS production. Neutrophils were analyzed by

flow cytometry using a Gallios Flow Cytometer (Beckman Coulter) and dihydrorhodamine 123

was used for detection of ROS.

We were unable to detect the presence of ROS in neutrophils stimulated with

complement anaphylatoxin C5a (Figure 6A). Stimulation with fMLP resulted in a 1.5 fold

increase in ROS production (Figure 6B), and stimulation with PMA resulted in approximately a

9 fold increase in ROS production (Figure 6C). These data suggest that activation of formylated

peptide receptor or PKC, respective targets for ligands fMLP and PMA, leads to neutrophil

oxidative burst, while activation of C5aR does not lead to oxidative burst. In this way,

neutrophils can modulate their response to activating stimuli. Recruitment of neutrophils to sites

of inflammation with anaphylatoxin C5a does not lead to neutrophil ROS production and

oxidative stress, while PKC activation, or stimulation of neutrophils with fMLP, can lead to ROS

production and oxidative stress.

Page 56: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

42

Figure 6. Neutrophil stimulation with fMLP and PMA, but not C5a, leads to oxidative

burst.

Neutrophils (1 × 106 cells/ml) were activated at 37°C for 30 min in the presence of

dihydrorhodamine 123 (10 μM) Cells were analyzed by flow cytometry on a Gallios Flow

Cytometer (Beckman Coulter). (A) Activation with C5a does not lead to oxidative burst.

Activation with (B) fMLP and (C) PMA leads to oxidative burst. Representative histogram is

shown. Results are given as median fluorescence intensity (MFI) from three independent

experiments. Student t-test, *P < 0.05, **p < 0.01, n=3.

Page 57: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

43

3.2.2 Complement Protein Localization Upon Activation

Neutrophil activation is a dynamic process where mobilization of neutrophil granules results in

exocytosis of proteins into the extracellular environment and modification of adhesion molecules

expressed on the plasma membrane. Upregulation of adhesion molecules enable neutrophils to

undergo their characteristic rolling, tethering, and transendothelial cell migration. In addition,

modulation of surface receptors allows for interaction with various ligands.

Neutrophil properdin has been shown to bind neutrophil surfaces and propagate

complement AP in a positive feedback loop for complement and neutrophil activation199

. In

addition, CFH can act as an adhesion protein and bind to integrin receptor CR3

(CD11b/CD18)206

. This suggests that activated neutrophils should bind complement proteins to

their surfaces.

To determine localization of the proteins previously identified (3.1), we utilized spinning-

disc immunofluoresecence confocal microscopy to analyze the cellular distribution of CFP (sc-

68366; Santa Cruz Biotechnology), C3 (ab97462; Abcam), CFB (sc-67151; Santa Cruz

Biotechnology), and CFH (AF4779; R&D Systems) upon activation with PMA (Figure 7). We

observed that upon neutrophil activation (Figure 7BDFH), complement proteins associate with

sialic acid surfaces, as indicated by an increase in colocalization between the complement

proteins and neutrophil sialic acids stained with wheat germ agglutinin.

During neutrophil activation, endogenous sialidases and sialyltransferases modulate the

expression of sialic acid on neutrophils and endothelial cells207

. Furthermore, neutrophil sialidase

activity on endothelial cell surfaces increases neutrophil adhesion and migration across

endothelium208

. Currently, it is unknown why complement proteins associate with sialic acid

upon activation, however this interaction suggests that activated neutrophils can potentially

mediate complement AP by modulating expression of sialic acids.

Page 58: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

44

Figure 7. Complement proteins associate with sialic acid upon activation.

Isolated neutrophils were left (ACEG) untreated or (BDFH) activated with PMA (20 nM) for 30

min. Cells were permeabilzed 0.2% (v/v) triton X-100 for 20 minutes. Samples were prepared

for immunostaining for detection of (AB) Properdin (sc-68366; Santa Cruz Biotechnology);

(CD) C3 (ab97462; Abcam); (EF) CFB (sc-67151; Santa Cruz Biotechnology), and (FG) CFH

(AF4779; R&D Systems). Neutrophil sialic acid was stained with Wheat Germ Agglutinin 568

(Invitrogen). Neutrophil nucleus was stained with DAPI. Z-stack images shown are

representative of images from three independent experiments taken with a 60×/1.35 oil

immersion objective. (ABGH) Scale bar, 3.60 μm. (CDEF) Scale Bar, 3.70 μm.

Page 59: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

45

3.2.3 Complement Protein Secretion in Response to Activation

Activation of neutrophils results in secretion of proteins stored in granules. Isolated neutrophils

(2 × 107

cells/ml) were activated with C5a (1 μM), fMLP (1 μM), and PMA (20 nM) to

determine if complement proteins are secreted. Neutrophil cell fractions were harvested through

centrifugation at 1000 × g and supernatants were collected for secreted proteins. Total protein

concentration was determined using a BCA assay, and 50 μg of protein was loaded onto SDS-

PAGE for western blot.

Activation of neutrophils with C5a, fMLP, and PMA led to secretion of complement

proteins (Figure 8). The absence of structural protein β-actin in the supernatant fractions

indicates that proteins collected are representative of secreted proteins. Activation of neutrophils

did not result in cell lysis which would release β-actin into the solution. CFP secretion from

activated neutrophils varied with response to different activating stimuli (Figure 8A). The

incremental increase in CFP secretion corresponds with the ROS production from the activating

stimuli (Figure 6). Activation of neutrophils, with PMA, resulted in the greatest concentration of

CFP secretion. Activation, with fMLP, resulted in lower concentrations of CFP secretion when

compared to PMA, and activation with C5a resulted in the least amount of CFP secretion. In

addition, CFP is retained in neutrophils even after secretion as indicated by the band found in the

lysate fraction of the western blot. Likewise, CFB also displayed a varying response of protein

secretion with an incremental increase corresponding to the ROS production of the activating

stimuli Figure 6. Activation with C5a and fMLP resulted in lower levels of CFB secretion, and

activation with PMA resulted in greater amounts of CFB secretion. We also noticed that only full

length CFB (90 kDa) is secreted into the supernatant while cleaved fragment CFB Bb (60 kDa) is

retained within the neutrophil (Figure 8C). C3 (Figure 8B) and CFH (Figure 8D) were

completely secreted into the supernatant upon activation with C5a, fMLP and PMA.

The varying secretion profiles for the complement proteins suggests that these proteins

are stored in different granules. The high propensity for mobilization of CFH and C3 suggest that

these proteins are likely to be stored in secretory granules92

. Neutrophil properdin is stored in

secondary granules198

, which have a much lower propensity for mobilization as demonstrated by

the varied response to activating stimuli209

. The secretion profile for CFB is similar to CFP

suggesting that they may be stored in the same granule. The varied secretion of complement

Page 60: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

46

proteins in response to activation suggests a role for neutrophils in modulating complement

activation. Early activation and secretion of CFH and C3 can serve to regulate complement

activity and modulate complement response, where as activation and mobilization of CFP and

CFB can greatly amplify the complement AP.

Page 61: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

47

Figure 8. Activated neutrophils secrete complement proteins.

Neutrophils were activated with different activating agents for 30 minutes. Cells were harvested

through centrifugation. Neutrophil lysates were prepared from the harvested cell pellet at a

concentration of 2 × 107 cells/ml. 50 μg of protein of lysate and equal volumes of supernatant, 50

μl, was loaded for detection of complement proteins. (A) CFP, 10% (w/v) non reducing SDS-

PAGE, 5 sec exposure on radiographic film. (B) C3, 10% (w/v) reducing SDS-PAGE, 5 min

exposure on radiographic film. (C) CFB, 10% (w/v) reducing SDS-PAGE, 5 min exposure on

radiographic film. (D) CFH, 10% (w/v) non-reducing SDS-PAGE, 5 min exposure on

radiographic film.

Page 62: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

48

3.3 Quantification of Nuclear Morphologies During NETosis

Activated neutrophils can generate NETs consisting of DNA and antimicrobial peptides to

effectively ensnare and kill bacteria 100

. To validate our method of producing NETs, we activated

neutrophils with PMA (20 nM) and fixed them with paraformaldehyde (4% w/v) at various time

points (0, 30, 60, 120, 180, 240 min). Cells were stained with DAPI and analyzed with

fluorescence microscopy (Figure 9) .

When neutrophils are activated to form NETs, four distinct nuclear morphologies can be

identified (Figure 9A). Resting neutrophils are multilobed (multilobed nuclei), however as the

genomic DNA begins to decondense in preparation for NETs, the nucleus begins to lose its

multilobed structure and appears to be delobulated (delobulated nuclei). As the nuclear envelope

disintegrates to allow mixing of DNA with neutrophil granule proteins, the nucleus begins to

look diffuse (decondensed nuclei). Finally, neutrophil DNA along with its antimicrobial peptides

are released to form NETs (Neutrophil Extracellular Traps).

Upon identifying four distinct nuclear morphologies, we examined the population of each

morphology during the course of NET formation along various time points. In our experiment,

the earliest time for detection of NETs was at 120 minutes, however the percentage difference

between NETs and other nuclear morphologies was not statistically significant. At 120 minutes,

the predominant nuclear morphology was the delobulated nucleus (p<0.0001) (Figure 9B). At

180, NETs were the predominant nuclear morphology (p<0.0001) (Figure 9B). The percentage

of nucleus found in the decondensed stage remains very low throughout neutrophil activation.

This suggests that this morphology is short lived and as the nuclear envelope disappears, nuclear

DNA rapidly mixes with cytosolic and granular proteins to form NETs. The differential cell

counts for nuclear morphology validates that our procedure for inducing NETosis in neutrophils

isolated from human peripheral whole blood is consistent with the methods described in

literature100,116

.

Page 63: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

49

Figure 9. Activated neutrophils form neutrophil extracellular traps.

Neutrophils were activated with PMA (20 nM) to induce the formation of NETs. Samples were

fixed with 4% (w/v) paraformaldehyde and stained with DAPI for microscopy. (A) Four distinct

nuclear morphologies (lobulated, delobulated, decondensed, NETs) can be identified during

NETosis. (B) Percentage difference for each nuclear morphology at various time points during

neutrophil activation. Percentage difference for nuclear morphologies was identified through

manual counting of at least 118 to 220 cells from 5 different focal planes at 40 × magnification.

Data are presented as mean ± SEM from 3 independent experiments. Statistical significance is

shown only if percentage of nuclear morphology is significant compared to all other

morphologies at the same time point. 2 way ANOVA with Tukey’s multiple comparison test, *p

<0.05, #p<0.001,

$p<0.0001.

Page 64: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

50

3.4 PMA, but not C5a and fMLP, Induce NET Formation

Neutrophils are recruited to sites of tissue (e.g. endothelium) injury and/or infection by

cytokines (e.g. IL-1β and TNF-α), growth factors (e.g. G-CSF), anaphylatoxins (e.g. C5a) and

other cell damage signals (e.g. fMLP). Upon arrival to sites of high chemoattractant

concentration, neutrophils coordinate a strict regiment to deal with potential threats through

programmed phagocytosis, degranulation, and NETosis.

To determine whether neutrophil activation with complement anaphylatoxin C5a, or

pathogen/damage signal fMLP, can lead to the formation of neutrophil extracellular traps, we

utilized a SYTOX green plate reader assay. SYTOX green is a cell impermeable DNA dye that is

excitable at 488 nm. It exhibits a 500-fold fluorescence enhancement upon binding to nucleic

acids. Using this assay, we were able to detect the presence of extracellular DNA as fluorescence

emission at 520 nm. The fluorescence was then used to quantify the formation of NETs.

Neutrophils (3 × 104) in RPMI media were seeded onto 96 well plates in the presence of

SYTOX green (5μM). Neutrophils were activated with C5a (1 μM, 2 μM), fMLP (1 μM, 2 μM)

and PMA (20 nm) and fluorescence (485/520) was taken at various time points to monitor the

formation of neutrophil extracellular traps. Our data indicates that neither C5a (Figure 10A), nor

fMLP (Figure 10B), were was able to induce the formation of NETs. Only activation with PMA

(Figure 10C) led to the formation of NETs.

The mechanism for NETosis is heavily dependent on the formation of ROS210

. Our

earlier data demonstrate that stimulation of neutrophils with C5a did not lead to an oxidative

burst and activation with fMLP only led to a 1.5 fold increase in ROS (Figure 6). In contrast,

stimulation of neutrophils with PMA leads to a 9 fold increase in production of ROS (Figure 6).

Thus, our data indicate that neutrophil response to C5a and fMLP does not lead to the formation

of NETs. This may be due to insufficient production of oxygen radicals.

Page 65: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

51

Figure 10. PMA, but not C5a and fMLP, induces NET formation.

Neutrophils (3 × 104) were seeded onto a 96 well tissue culture dish in the presence of SYTOX

green (5μM). NET formation was inhibited with addition of NOX inhibitor DPI (2μM). (A) C5a

(1μM or 2μM) activation of neutrophils did not lead to the formation of NETs. Data are

presented as mean ± SEM from 3 to 7 individual experiments. (B) fMLP (1μM or 2μM)

activation of neutrophils did not lead to the formation of NETs. Data are presented as mean ±

SEM from 3 to 7 individual experiments. (C) PMA (20 nM) activation of neutrophils led to the

formation of NETs. Data are presented as mean ± SEM from 7 individual experiments.

Fluorescence emission of SYTOX green was normalized to total DNA of resting neutrophils

permeabilized with 0.5% triton X-100. Two-way ANOVA with Tukey’s multiple comparison

test, $P < 0.0001.

Page 66: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

52

3.5 Neutrophil Extracellular Traps Contain Properdin

Although not exclusive, the association of proteins to NETs can be associated with charge

interactions133

. Complement regulatory proteins, CFP and CFH, have been shown to associate

with polyanionic compounds23,211

. Moreover, our previous data suggest that C3 and CFB

associate with sialic acid upon neutrophil activation (Figure 7). Taken together, these data

strongly suggest that complement proteins may associate with polyanionic structures such as

neutrophil extracellular traps.

To determine if the complement AP proteins contained in neutrophils are present in

NETs, we isolated neutrophils from human peripheral whole blood and prepared them for

spinning-disc immunofluorescence confocal microscopy. Neutrophils were seeded onto 12 mm

coverslips, coated with poly-L-lysine, and activated with PMA (20 nM). After 240 minutes of

activation, neutrophils were fixed with 4% (w/v) paraformaldehyde. Cells were not

permeabilized because we were only interested in extracellular structures. Samples were stained

for CFP (sc-68366; Santa Cruz Biotechnology) (Figure 11), C3 (ab97462; Abcam) (Figure 12),

CFB (sc-57151; Santa Cruz Biotechnology) (Figure 13), and CFH (AF4779; R&D Systems)

(Figure 14). Samples were co stained with myeloperoxidase (ab25989; Abcam), a protein which

is known to be found in NETs. Our data indicates that CFP (Figure 11) can be found in NETs, as

it follows same pattern of distribution as neutrophil MPO and extracellular DNA stained with

DAPI. C3 (Figure 12), CFB (Figure 13) and CFH (Figure 14) were not found on NETs. From

these data, we can conclude that neutrophil properdin is associated with neutrophil extracellular

traps.

However, with these current data, we cannot rule out the possibility that other

complement proteins may also be present in NETs. We previously identified the concentration of

CFP to be much higher in neutrophils than the concentration of C3, CFB and CFH (Figure 5).

The concentration of these proteins may be below the limit of detection for immunofluorescence

microscopy. This is further complicated because attempts to detect these proteins through

increased exposure of fluorophore to the microscope laser or increasing detection sensitivity of

the camera can lead to autofluorescence in neutrophils 212-214

. Other more sensitive techniques

(e.g. ELISA) maybe required to definitively determine whether neutrophil complement proteins

are found in NETs. Our current data only demonstrate that CFP is associated with NETs.

Page 67: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

53

Figure 11. Neutrophil extracellular traps contain properdin.

Neutrophil extracellular traps are prepared for spinning-disc immunofluorescence microscopy.

Properdin is found in secondary granules in resting neutrophils. In order to detect only properdin

on NETs, samples were not permeabilized. (A) Properdin was detected using secondary donkey

anti-rabbit Alexa Fluor ® 555 antibody. (B) Myeloperoxidase was detected using secondary

donkey anti-mouse Alexa Fluor ® 488 antibody. (C) Neutrophil extracellular traps were detected

with DAPI. (D) CFP and MPO colocalize to neutrophil extracellular traps. Z-stack images shown

are representative of images from five independent experiments taken with a 60×/1.35 oil

immersion objective. Scale bar, 9.00 μm.

Page 68: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

54

Figure 12. C3 does not interact with neutrophil extracellular traps.

Neutrophil extracellular traps are prepared for spinning-disc immunofluorescence microscopy.

Samples are not permeabilized in order to detect protein associated with extracellular DNA. (A)

C3 was detected using secondary donkey anti-rabbit Alexa Fluor ® 555 antibody. (B)

Myeloperoxidase was detected using secondary donkey anti-mouse Alexa Fluor ® 488 antibody.

(C) Neutrophil extracellular traps were detected with DAPI. (D) C3 immunostain does not

associate with MPO or extracellular DNA. Z-stack images shown are representative of images

from five independent experiments taken with a 60×/1.35 oil immersion objective. Scale bar,

9.00μm

Page 69: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

55

Figure 13. CFB does not interact with neutrophil extracellular traps.

Neutrophil extracellular traps are prepared for spinning-disc immunofluorescence microscopy.

Samples were not permeabilized in order to detect protein associated with extracellular DNA.

(A) CFB was detected using secondary donkey anti-rabbit Alexa Fluor ® 555 antibody. (B)

Myeloperoxidase was detected using secondary donkey anti-mouse Alexa Fluor ® 488 antibody.

(C) Neutrophil extracellular traps were detected with DAPI. (D) CFB does not appear to

associate with MPO or extracellular DNA. Z-stack images shown are representative of images

from five independent experiments taken with a 60×/1.35 oil immersion objective. Scale bar,

9.00μm

Page 70: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

56

Figure 14. CFH does not interact with neutrophil extracellular traps.

Neutrophil extracellular traps are prepared for spinning-disc immunofluorescence microscopy.

Samples were not permeabilized in order to detect protein associated with extracellular DNA.

(A) CFH was detected using secondary donkey anti-goat Alexa Fluor ® 555 antibody. (B)

Myeloperoxidase was detected using secondary donkey anti-mouse Alexa Fluor ® 488 antibody.

(C) Neutrophil extracellular traps were detected with DAPI. (D) CFH immunostain does not

associate with MPO or extracellular DNA. Z-stack images shown are representative of images

from five independent experiments taken with a 60×/1.35 oil immersion objective. Scale bar,

9.00μm

Page 71: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

57

3.6 NETs Increase Formation of Terminal Complement Complex

The presence of properdin on neutrophil extracellular traps suggests that NETs may activate

complement through the complement alternative pathway23

. Although there are several pathways

for complement initiation and activation, all pathways converge to the formation of the terminal

complement complex (C5b-9). Thus, complement activation can readily be detected through

formation of C5b-9.

To determine the effect of NETs on complement activation, neutrophils were isolated and

activated with PMA (20 nM) for 240 minutes in RPMI1640 supplemented with 10 mM Hepes.

After activation, neutrophils were centrifuged at 200 × g for 5 minutes and RPMI media was

replaced with 20% (v/v) sterile filtered autologous FFP in RPMI media. Complement competent

plasma was prepared with Refludan ®, a potent thrombin inhibitor to prevent coagulation. Since,

complement CP is dependent on Ca2+

and complement AP is dependent on Mg2+

, the use of

chelating agents as anticoagulants, such as EDTA, will inhibit complement activation and

therefore cannot be used to prepare complement competent plasma. After addition of 20% (v/v)

autologous FFP:RPMI, neutrophils were incubated at 37°C, 5% (v/v) CO2 for 15 minutes to

allow for the formation of C5b-9 and fixed with 4% (w/v) paraformaldehyde for spinning-disc

immunofluorescence confocal microscopy. In the rabbit hemolysis assay (Appendix B),

incubation of rabbit erythrocytes with 20% complement competent plasma leads to 67 to 100%

hemolysis (Table B2). Thus, complement activation is a rapid process and incubation of samples

with complement competent plasma for 15 minutes is sufficient for complement activation and

formation of C5b-9.

Our data indicate that addition of plasma did not initiate the formation of C5b-9 in non

stimulated neutrophils (Figure 15ADG). In the presence of NETs, the addition of complement

competent plasma led to the formation of C5b-9 (Figure 15BEH). This effect was abolished

when neutrophils were activated in the presence of DNase I (50 μg/ml) before the addition of

complement competent plasma (Figure 15CFI). Together, these data indicate that NETs can

activate complement pathways which result in the formation of C5b-9.

Page 72: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

58

Figure 15. Neutrophil extracellular traps increase formation of C5b-9. Neutrophils were left untreated (ADG) activated with 20 nM PMA (BEH) and treated with

DNase I to disseminate the formation of NETs (CFI). After induction of NETs, 20% (v/v) sterile

filtered autologous FFP was added to samples. (A-C) NETs were visualized with DAPI, (D-F)

C5b-9 was detected using mouse monoclonal antibody to C5b-9 (DIA 011-01; Antibody shop).

Representative image from three independent experiments taken with 60×/1.35 oil immersion

objective is shown. Data are presented in extended focus. Scale bar, 9.00μm.

Page 73: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

59

3.7 NETs Activate Complement Alternative Pathway to Form C5b-9

The presence of NETs is able to initiate complement activation to form C5b-9 (Figure 15).

However, the presence of complement AP proteins identified in neutrophils (Figure 5) strongly

suggests that complement amplification may be through complement AP.

In order to determine if NETs can activate specifically the complement AP, we utilized a

complement AP buffer, containing 20 mM Hepes, pH 7.4, 144 mM NaCl, 7 mM MgCl2, and 10

mM EGTA. EGTA is a chelating agent with low affinity for Mg2+

making it selective for Ca2+

.

The presence of EGTA inhibits complement CP, while the addition of Mg2+

enables the

activation of complement AP.

Neutrophils were activated with PMA (20 nM) for 240 minutes in RPMI1640

supplemented with 10 mM Hepes. After activation, neutrophils were centrifuged at 200 × g for 5

minutes and a buffer exchange was performed through three washes with PBS followed by one

wash with AP buffer. 20% (v/v) sterile filtered autologous FFP in AP buffer was added to each

sample and neutrophils were incubated at 37°C, 5% CO2 for 15 minutes to allow for the

formation of C5b-9. Samples were fixed with 4% (w/v) paraformaldehyde and prepared for

spinning-disc immunofluorescence confocal microscopy.

Our data indicate that addition of plasma in AP buffer did not initiate the formation of

C5b-9 in non stimulated neutrophils (Figure 16ADG). In the presence of NETs, the addition of

20% (v/v) plasma in AP buffer led to the formation of C5b-9 (Figure 16BEH). The pattern of

C5b-9 formation detected in AP buffer (Figure 16) varies with the pattern detected in RPMI

media (Figure 15). Complement activation is dependent on the presence of divalent cations Ca2+

and Mg2+

. RPMI media (Wisent Bioproducts, Montreal, QC, Canada) contains 400 μM of Ca2+

and Mg2+

. In contrast, AP buffer contains 7 mM of Mg2+

. In a buffer designed for complement

activation, we see a greater formation of C5b-9. This effect was abolished when neutrophils were

activated in the presence of DNase I to digest extracellular DNA (50 μg/ml) before the addition

of plasma in AP buffer (Figure 16CFI). Together, these data indicate that NETs can activate

complement through the alternative pathway to form C5b-9.

Page 74: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

60

Figure 16. NETs activate complement alternative pathway to form C5b-9 Neutrophils were left untreated (ADG) activated with 20 nM PMA (BEH) and treated with

DNase I to disseminate the formation of NETs (CFI). A buffer exchange was performed with

three washes of PBS and one wash of AP buffer, followed by addition of 20% (v/v) autologous

plasma in AP buffer. (A-C) NETs were visualized with DAPI, (D-F) C5b-9 was detected using

mouse monoclonal antibody to C5b-9 (DIA 011-01; Antibody shop). Image was taken with

60×/1.35 oil immersion objective. Data are presented in extended focus. Scale bar, 9.00μm.

Page 75: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

61

3.8 NETs Activate Complement and Activation of Complement AP on NETs is Dependent on Properdin

Properdin is the major positive regulator of complement AP. Blocking of properdin can inhibit

the amplification of complement AP without affecting other modes of complement

amplification215

. To investigate if other modes of complement activation are activated in NETs,

we utilized a mouse monoclonal antibody to properdin (A233; Quidel Corporation) that is

capable of blocking its activity215

. We performed a serial dilution of the mouse monoclonal

antibody, and complement activity was determined through rabbit erythrocyte hemolysis assay.

Rabbit erythrocytes do not contain sialic acid therefore are unable to bind CFH. In addition, they

lack the membrane bound regulators for complement AP, thus the constitutively active

complement AP is able to propagate on rabbit erythrocyte surface to form C5b-9 leading to cell

lysis. Complement activity is determined through measuring free hemoglobin in supernatants

with end point absorption at 405 nm.

The concentration of antibody required to inhibit the activation complement AP in 20%

(v/v) plasma varied from donor to donor. In three independent donors, we determined the

effective range varied from 2-8 μg/ml. The data presented correspond to 20% (v/v) plasma that

showed effective inhibition of complement AP from 4μg/ml of blocking antibody (Figure 17A).

To examine whether NETs activate other modes of complement amplification, neutrophils were

activated with PMA (20 nM) for 240 minutes to induce the formation of NETs. After induction

of NETosis, a buffer exchange is performed and neutrophil media is replaced with 20% (v/v)

autologous FFP in AP buffer or 20% (v/v) plasma in RPMI1650 supplemented with 10 mM

Hepes. Both these buffers are incubated in the presence or absence of 4 μg/ml of the blocking

antibody to CFP.

Our data reveal that NETs are able to propagate the assembly of C5b-9 when

supplemented with 20% (v/v) autologous FFP in AP buffer (Figure 17B). However inhibition of

CFP is able to stop the complement activation and C5b-9 formation (Figure 17C). NETs

supplemented with 20% (v/v) autologous FFP in RPMI are also able to propagate complement

activation to form C5b-9 (Figure 17D), however addition of mAb to CFP in RPMI does not

inhibit C5b-9 formation (Figure 17E).

Page 76: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

62

Together, these data indicate that activation of complement AP on NETs is dependent on

properdin. However, inhibition of properdin had no effect on complement activation in RPMI

media, thus revealing that NETs activate complement through other modes of complement

activation in addition to complement AP.

Page 77: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

63

Figure 17. Properdin is required for activation of complement AP on NETs. Mouse monoclonal antibody to properdin inhibits the activation of complement alternative

pathway on NETs. (A) Serial dilution is performed to determine the concentration of mouse

mAb to CFP required to inhibit complement AP as determined by rabbit erythrocyte hemolysis.

Neutrophils are activated with PMA (20 nM) to induce the formation of NETs. A buffer

exchange was performed followed by addition of (B) 20% (v/v) plasma:AP buffer – mAb CFP,

(C) 20% (v/v) plasma:AP buffer + mAb CFP, (D) 20% (v/v) plasma:RPMI1640 + 10 mM hepes

– mAb CFP, (E) 20% (v/v) plasma:RPMI1640 + 10 mM Hepes + mAb CFP. Image was taken

with 60×/1.35 oil immersion objective. Data are presented in extended focus. Scale bar, 9.00μm.

Page 78: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

64

4 Results – Aim 2

To determine the significance of complement-NET interactions in antimicrobial function.

4.1 Identifying Bacteria That Induce NETosis

In order to determine the significance of neutrophil-complement interactions with regards to

antimicrobial function, we set out to identify bacteria, which can activate neutrophils to induce

the formation of neutrophil extracellular traps. Using the SYTOX green plate reader assay, we

activated neutrophils with a panel of different bacteria to examine their roles in activating

NETosis.

From our selected panel of bacteria, we identified that activation of neutrophils with

Pseudomonas aeruginosa (Figure 18) and Staphylococcus aureus (Figure 19A) led to the

formation of NETs. We had access to three different strains of P. aeruginosa. All three strains

mPAO1 (Figure 18A), PAKwt (Figure 18B), and PAKgfp (Figure 18C) led to the formation of

NETs and displayed similar kinetics to PMA activated NETs. Moreover, NET formation is

concentration dependent with regards to the number of bacteria used to activate neutrophils.

With all three strains, activation with a multiplicity of infection of 100 (MOI of 100) led to an

increase in NETs when compared with activation with a multiplicity of infection of 10 (MOI of

10).

Activation of neutrophils with S. aureus (RN6390), led formation of neutrophil

extracellular traps (Figure 19A). The formation of NETs in response to S. aureus was also

concentration dependent. Activation of neutrophils with MOI of 100 led to an increase in NET

production when compared to activation with MOI of 10. Activation of neutrophils with S.

aureus at MOI 100 led to a robust NET formation with SYTOX fluorescence being detected at

levels greater than total neutrophil DNA. Moreover, S.aureus activation using a MOI of 100

could not be completely inhibited with NOX inhibitor DPI, suggesting that the formation of

NETs in response to S. aureus has both ROS dependent and independent mechanisms.

Previous reports demonstrated that activation of neutrophils with bacterial

lipopolysaccharide (LPS) can lead to the formation of NETs100

. Interestingly, our data showed

that activation of neutrophils with gram-negative Escherichia coli (Y1088) at MOI 10 does lead

to the formation of NETs. The neutrophil response to E. coli is concentration dependent as

Page 79: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

65

activation with a MOI of 100 led to the formation of NETs. There is a delayed onset for the

release of extracellular traps when neutrophils are activated with E. coli at a MOI of 100,

however there is a rapid increase of extracellular DNA at 180 minutes (Figure 19B).

Activation of neutrophils with Gram-positive Bacillus subtilis was also concentration

dependent. Activation with B. subtilis at a MOI of 10 did not lead to the formation of NETs.

Activation with B. subtilis at MOI 100 led to the formation of neutrophil extracellular traps

(Figure 19C).

Together, these data indicate that there are other factors outside of Gram-classification

which can influence neutrophils to form NETs in response to bacteria. LPS alone may not be

sufficient to induce the formation of NETs, as Gram-negative E. coli are poor inducers of

NETosis. In addition, bacterial LPS may not be required to induce formation of NETs, as Gram-

positive S. aureus is a strong inducer of NETosis. The neutrophil response to different virulent

factors may also play a role in triggering NETosis, for example, pyocyanin is a virulent factor

associated with P. aeruginosa and has been demonstrated to activate NET formation in an

NADPH dependent manner143

. Other virulent factors specific for activating NETosis have yet to

be identified. It may be possible that more virulent strains of otherwise harmless bacteria (e.g. E.

coli O157:H7) may possess virulent factors which can potentially trigger NETosis. Thus, our

current findings can only be applied to these specific strains of bacteria tested.

Page 80: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

66

Figure 18. Pseudomonas aeruginosa activate neutrophils to form NETs

Neutrophils (3 × 104) in RPMI were seeded onto a 96 well tissue culture dish in the presence of

SYTOX green (5μM). NET formation was inhibited with addition of NOX2 inhibitor DPI

(2μM). Neutrophils were activated with different strains of Pseudomonas aeruginosa.(A) P.

aeruginosa, mPAO1. Data are presented as mean ± SEM from 5 to 7 individual experiments. (B)

P. aeruginosa, PAKwt. Data are presented as mean ± SEM from 5 to 7 individual experiments.

(C) P. aeruginosa, PAKgfp. Data are presented as mean ± SEM from 4 to 7 individual

experiments. Fluorescence emission of SYTOX green was normalized to total DNA of resting

neutrophils permeabilized with 0.5% triton X-100. Statistical significance compared to SYTOX

fluorescence from resting neutrophils. 1denotes statistical significance between different

multiplicities of infection. 2 way ANOVA with Tukey’s multiple comparison test, *p <0.05,

**p<0.01, #p<0.001,

$p<0.0001.

Page 81: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

67

Figure 19. Neutrophils activated with S. aureus, E. coli, and B. subtilis.

Neutrophils (3 × 104) in RPMI were seeded onto a 96 well tissue culture dish in the presence of

SYTOX green (5μM). NOX2 dependent NETosis was inhibited with DPI (2μM). (A) S. aureus,

RN6390. Data are presented as mean ± SEM from 4 to 7 individual experiments. (B) E. coli,

Y1088. Data are presented as mean ± SEM from 5 to 7 individual experiments. (C) B. subtilis,

unknown strain. Data are presented as mean ± SEM from 4 to 7 individual experiments.

Fluorescence emission of SYTOX green was normalized to total DNA of resting neutrophils

permeabilized with 0.5% triton X-100. Statistical significance compared to SYTOX fluorescence

from resting neutrophils. 1 denotes statistical significance between different multiplicities of

infection. 2 way ANOVA with Tukey’s multiple comparison test, *p <0.05, **p<0.01, #p<0.001,

$p<0.0001.

Page 82: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

68

4.2 Neutrophil Properdin Binds P. aeruginosa Independent of NETs

In the previous chapter, we demonstrated that neutrophil properdin can be found on NETs

(Section 3.5). To determine if neutrophils can target CFP onto bacteria through NETs, we

utilized a transgenic strain of pseudomonas, transformed using a mini-Tn7 transposon delivery

system. This chromosomally labeled P. aeruginosa (PAKgfp) expresses gfp under a

constitutively active promoter 216

.

Our data indicate that neutrophil properdin binds to Pseudomonas aeruginosa PAKgfp

(Figure 20). However, this was independent of neutrophil NET formation. Activation of

neutrophils with PAKgfp for 30 minutes (Figure 20ADGJ) led to CFP deposition onto bacteria.

Neutrophil activation at 30 minutes shows secretion of CFP in response to formylated peptides,

however, no NETs are formed at this time (Figure 8). Activation of neutrophils to induce the

formation of NETs also led to deposition of CFP onto bacteria (Figure 20BEHK). Addition of

DNase I to disseminate NETs did not result in decreased deposition of CFP to PAKgfp (Figure

20CFIL).

Statistical analysis of thresholded Pearson’s correlation between neutrophil properdin and

PAKgfp confirmed that properdin binds bacteria independent of NETs. Colocalization data were

obtained from 40 individually selected bacteria from 3 focal planes and average thresholded

pearson’s correlation from 3 independent experiments ± SEM was calculated. The colocalization

of neutrophil CFP to PAKgfp after 30 minutes of activation was 0.801 ± 0.038, the

colocalization of neutrophil CFP to PAKgfp in the presence of NETs was 0.740 ± 0.007, and

colocalization of neutrophil CFP to PAKgfp in the presence of DNase I was 0.779 ± 0.014.

Statistical analysis using 2 way ANOVA with Tukey’s multiple comparison test reported no

statistical significance between these values. Therefore, while neutrophil CFP is capable of

binding to P. aeruginosa, the formation of NETs does not actively target P. aeruginosa to

deposit CFP onto bacteria surfaces.

Page 83: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

69

Figure 20. Properdin binds P. aeruginosa independent of NETs.

Neutrophils were activated with PAKgfp for 30 min (ADGJ), 240 min (BEHK) and 240 min

with DNase I (CFIL). Neutrophil properdin (D-F) binds to PAKgfp (G-I). Merged images (J-L)

with Pearson’s thresholded correlation ± SEM from three independent experiments. Z-stack

images shown are representative of images from three independent experiments taken with a

60×/1.35 oil immersion objective. Scale bar, 9.00μm.

Page 84: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

70

4.3 NETs Increase Formation of C5b-9 in Response to Bacteria

In the previous chapter (Section 3.6) we demonstrated that NETs increase complement activation

and lead to the formation of the membrane attack complex. Although CFP was not targeted to P.

aeruginosa via NETs, the activation of complement on bacteria trapped by NETs can still be an

effective means of microbial clearance.

Neutrophils were activated with PAKgfp in the absence or presence of DNase I for 240

minutes. After activation, neutrophils were supplemented with 20% sterile autologous FFP and

incubated for an additional 15 minutes at 37°C, 5% CO2 to allow for the assembly of C5b-9.

Samples were fixed with 4% (w/v) paraformaldehyde for spinning-disc immunofluorescence

confocal microscopy.

We demonstrated that supplementing neutrophils with complement competent plasma,

did not initiate to the formation of C5b-9 in resting neutrophils that were not activated with P.

aeruginosa PAKgfp (Figure 21ADGJ). In the presence of NETs, the addition of complement

competent plasma led to the formation of C5b-9 (Figure 21BEHK). The formation of C5b-9 was

abolished when neutrophils were activated in the presence of DNase I (50 μg/ml), to disseminate

extracellular DNA, before the addition of complement competent plasma (Figure 21CFIL).

Together, these data indicate that NETs enhance complement amplification on trapped bacteria.

Page 85: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

71

Figure 21. P. aeruginosa activation of neutrophils increases C5b-9 formation on NETs.

Neutrophils untreated (ADGJ), activated with PAKgfp (BEHK) in the presence of DNase I

(CFIL). Addition of 20% (v/v) autologous FFP to NETs (A-C) leads to increased deposition of

C5b-9 (D-F) onto PAKgfp (G-I) activated neutrophils. Representative image from three

independent experiments taken with 60×/1.35 oil immersion objective is shown. Data are

presented in extended focus. Scale bar, 9.00 μm.

Page 86: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

72

4.4 NETs Activate Complement Alternative Pathway in Response to Bacteria

Complement activation can be initiated through several pathways, all of which converge to form

the terminal complement complex (C5b-9). NETs, which ensnare bacteria, are able to initiate the

complement response which results in the formation of C5b-9 (Figure 21). The abundance of

CFP in neutrophils suggests that they may be key mediators of the complement AP. The

dependency of the different complement pathways on different cations allows us to carefully

dissect which pathways of complement are activated to form C5b-9. Complement CP is

dependent on Ca2+

while complement AP is dependent on Mg2+

.

To determine if NETs can activate complement AP in response to P. aeruginosa, we

activated neutrophils with PAKgfp at a MOI of 10 to induce NETosis and performed a buffer

exchange. 20% (v/v) sterile filtered autologous FFP in AP buffer was added to each sample and

neutrophils were incubated at 37°C, 5% CO2 for 15 minutes to allow for the formation of C5b-9.

Samples were fixed with 4% (w/v) paraformaldehyde and prepared for spinning-disc

immunofluorescence confocal microscopy.

Our data indicate that addition of plasma in AP buffer does not initiate the formation of

C5b-9 in non stimulated neutrophils (Figure 22ADGJ). In the presence of NETs, the addition of

20% (v/v) plasma in AP buffer lead to the formation of C5b-9 (Figure 22BEHK). The ability of

NETs to propagate the formation of C5b-9 was abolished when neutrophils were activated in the

presence DNase I (50 μg/ml) before the addition of plasma in AP buffer (Figure 22CFIL).

Together, these data indicate that NETs formed in response to bacteria are effective at activating

complement AP. This suggests that NETs enhance complement amplification on ensnared

bacteria.

Page 87: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

73

Figure 22. NETs activate complement AP to form C5b-9 in presence of P. aeruginosa.

Neutrophils were left untreated (ADGJ), activated with PAKgfp (BEHK) and treated with DNase

I (CFIL). A buffer exchanged was performed, followed by addition of 20% (v/v) autologous

plasma in AP buffer. NETs were visualized with (A-C) DAPI, and immunostained for (D-F)

C5b-9, and (G-I) GFP. Image was taken with 60×/1.35 oil immersion objective. Data are

presented in extended focus. Scale bar, 9.00 μm.

Page 88: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

74

4.5 NETs Activate Complement in Response to P. aeruginosa and Complement AP Activation is Dependent on Properdin

Neutrophil extracellular traps formed in response to PMA are able to activate multiple

complement pathways (Section 3.8). As a mechanism for antimicrobial activity, NETs are very

potent at ensnaring invading bacteria. The ability to initiate multiple pathways of complement

activation on bacteria trapped in NETs could be a potent mechanism for microbial clearance.

To examine whether NETs can activate other pathways of complement activation in

response to P. aeruginosa, we utilized a mouse monoclonal antibody to properdin (A233; Quidel

Corporation) that is capable of blocking its activity215

. We used concentrations of this antibody

previously established to be effective in blocking complement AP activity (Figure 17A).

Neutrophils were activated with PAKgfp at a MOI of 10 for 240 min to induce the formation of

NETs. Following activation, a buffer exchange was performed and neutrophil media was

replaced with 20% (v/v) autologous FFP in AP buffer or 20% (v/v) plasma in RPMI1650

supplemented with 10 mM Hepes. Both these buffers are incubated in the presence or absence of

the mouse monoclonal antibody to properdin (A233; Quidel Corporation).

Neutrophils supplemented with 20% (v/v) autologous FFP in AP buffer are able to

propagate the assembly of C5b-9 in response to PAKgfp (Figure 23A). However, the formation

of C5b-9 in AP buffer was inhibited by the addition of the mouse monoclonal antibody to CFP

(Figure 23B). This suggests that complement AP activation on NETs in response to PAKgfp is

dependent on CFP. The addition of 20% (v/v) autologous FFP in RPMI to neutrophils was also

able to propagate the assembly of C5b-9 (Figure 23C), however the addition of mouse

monoclonal antibody to properdin did not block the assembly of C5b-9 (Figure 23D). This

suggests that in addition to complement AP, NETs are able to initiate the assembly of C5b-9

through other pathways of complement activation.

Together, these data indicate that activation of complement AP on NETs in response to P.

aeruginosa is dependent on properdin. However, inhibition of CFP had no effect on complement

activation in RPMI media, revealing that, in addition to complement AP, NETs activate

complement through other pathways in response to P. aeruginosa. The ability for NETs to

activate complement on bacteria may contribute to antimicrobial clearance.

Page 89: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

75

Figure 23. NETs activation of complement AP in presence of P. aeruginosa is dependent on

Properdin.

Addition of mAb to properdin stops activation of complement AP on NETs. Neutrophils are

activated with PAKgfp at a MOI of 10. A buffer exchange was performed followed by addition

of (A-B) 20% (v/v) plasma:AP buffer, or (C-D) RPMI1640 + 10 mM Hepes; (AC) without mAb

to properdin, or (BD) with addition of mAb to Properdin. Images were taken with 60×/1.35 oil

immersion objective. Data are presented in extended focus. Scale bar, 9.00 μm.

Page 90: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

76

5 Discussion

Failure to regulate the immune response can often lead to deleterious inflammatory damage

associated with neutrophil influx and complement activation. While evidence highlights

extensive cross-talk between neutrophil and complement activation, little is known with regards

to NETs. The goal of this work was to characterize the interactions between the complement

system and NETs in order to shed light on how the link between neutrophils and innate immunity

can contribute to disease pathology. For this we conducted ex vivo studies using neutrophils

purified from human periphal whole blood. We were able to demonstrate that, neutrophils, in

response to certain stimuli, can activate to form NETs which are able to propagate complement

activation to form C5b-9. The activation of complement AP on NETs is dependent on CFP,

however NETs are also able to initiate other pathways of complement activation that are

independent of CFP to form C5b-9. This suggests that NETs are able to amplify complement

activation through multiple pathways.

5.1 Characterizing Interactions of Complement and Neutrophils

We first identified complement proteins that are expressed in purified neutrophils. We found that

under resting conditions, neutrophils contain CFP, C3, CFB and CFH (Figure 5). Neutrophils are a

potent source of CFP in circulation. They synthesize CFP and store the protein in secondary granules

for release upon activation198. As a result, CFP was the most abundant protein of the four

complement proteins we identified in neutrophils. Consistent with literature, we also found that

neutrophils express C3 and CFB, although these proteins were found at much lower

concentrations198,217. CFP, C3 and CFB all play an important role in amplifying complement AP.

Certainly, in the presence of circulating CFD (1-8 μg/ml), neutrophils express all the necessary

constituents to activate complement through the AP205. In addition, we were able to detect expression

of CFH, the major soluble regulator of complement AP, in resting neutrophils. To our knowledge this

is the first time anyone has shown expression of CFH in neutrophil. The role neutrophil CFH plays in

the immune response is currently unknown. It may act as an adhesion molecule for phagocytosis, as

binding of CFH to CR3 mediates attachment to Candida albicans to enhance neutrophil

antimicrobial activity194. The identification of these proteins in neutrophils, which are intricately

involved in complement AP activation and regulation, suggest that they play a prominent role in

activating complement AP.

Page 91: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

77

In order to gain a better understanding of how these complement proteins interact with

neutrophils, we decided to investigate the neutrophil response to various activating stimuli. We

selected complement C5a, a powerful anaphylatoxin, fMLP, which can serve as both a PAMP

and a DAMP, and PMA, a potent PKC activator. Neutrophil activation is associated with an

oxidative burst to produce ROS. Using flow cytometry, we measured neutrophil ROS production

using DHR123. Our findings indicate that activation with fMLP leads to mild oxidative burst and

activation with PMA leads to a dramatic production of ROS, however activation of neutrophil

with C5a is not sufficient to induce respiratory burst (Figure 6). This is consistent with the

literature as C5a has a priming effect on neutrophils. Activation with C5a alone is not sufficient to

induce oxidative burst as measured with DHR123218. However, priming of neutrophils enhances its

ability to produce reactive oxygen metabolites in response to activating stimuli219,220. Therefore, in

the presence of complement amplification and c5a anaphylatoxin production, neutrophils maybe

primed to more readily form ROS and undergo NETosis.

Another hallmark of neutrophil activation is granule mobilization and degranulation. To

further characterize neutrophil complement interactions in response to activation, we looked for

secretion of the previously identified complement proteins. We found that CFP and CFB secretion

varied with response to different activating stimuli, while CFH and C3 were completely secreted into

the supernatant (Figure 8). This sheds light on the granular localization of complement proteins

within neutrophils. As a reservoir for surface receptors, secretory vesicles have a low threshold for

mobilization and are completely mobilized to the surface upon activation in response to chemokines

(e.g. fMLP)92. The complete mobilization of CFH and C3 in response to all three of the activating

stimuli suggest that they are found in neutrophil secretory granules. This class of granule is relatively

new and a complete characterization of protein contents in the secretory vesicle has not yet been

performed95. Tertiary granules have a higher threshold for mobilization (20% mobilized by

fMLP)209,221, secondary granules have an even higher threshold for mobilization and primary

granules are often only mobilized partially88,209. CFP is a neutrophil protein known to be found in

secondary granules198. As a result, it is only partially mobilized in response to activation with C5a

and fMLP. The localization of CFB in neutrophils is currently unknown, however the varied

response of CFB secretion suggests that it is in one of these 3 other classes of granules that are

formed earlier during neutrophil development. The varying thresholds for granule mobilization

serves as a way for regulating neutrophil activation. In this way, mobilization of secretory vesicles

can decorate neutrophil plasma membrane with receptors vital for immune response without release

Page 92: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

78

of harmful granule proteins95. Secretion of C3 and CFH at a much lower threshold suggest roles for

neutrophil in regulating complement response and opsonization without formation of convertases and

C5b-9. Secretion of CFB and CFP from neutrophils represents an efficient means for complement AP

amplification.

5.2 Properdin is Found on Neutrophil Extracellular Traps

To further characterize the interaction between complement system in neutrophils, we looked at

NETs. Using a SYTOX green plate reader assay, we found that neither C5a or fMLP alone was

sufficient to induce the process of NETosis (Figure 10). This indicates that complement

activation to produce anaphylatoxins or formylated peptides alone are not sufficient to trigger

neutrophils to form NETs. C5a however may play an important role in neutrophil priming to

enhance the process of NETosis. Consistent with literature, activation of neutrophils with PMA

triggered a robust oxidative burst (Figure 6) and formation of NETs (Figure 10)100.

Using PMA as a model for NETosis, we set forth to characterize the interaction of CFP, C3,

CFB and CFH with NETs. In 2009, mass spectrometry analysis identified 24 proteins to be

associated with NETs109. The complement proteins we identified were not present in this study.

However, complement proteins are often over looked in mass spectrometry analysis as they are often

associated with plasma proteins. Our findings indicate that CFP can be found on NETs (Figure 11).

C3 (Figure 12), CFB (Figure 13) and CFH (Figure 14), were not identified on NETs. However, our

findings suggest that the concentration of CFP in neutrophils is much greater than C3, CFB or CFH

(Figure 5). The concentration of these proteins dispersed in NETs may be below the limit of

detection for immunofluorescence microscopy. Attempts to detect these proteins by increasing

exposure of the fluorophore to the microscope laser or increasing detection sensitivity of the camera

can lead to autofluorescence in neutrophils 212-214

. Thus at this time, we are unable to conclude that

these proteins are not associated with NETs.

The mechanism for deposition of CFP onto NETs may in part be due to its pattern

recognition molecule properties. As a known pattern recognition molecule, CFP has been reported to

be able to bind to necrotic cells via surface DNA31. In circulation, CFP is a multimeric protein that

forms dimers, trimers and tetramers in a head to tail fashion33. The monomer is a 53 kDa protein

composed exclusively of 7 thrombospondin repeats (TSR)33,34. TSRs contain two arginine residues

which are thought to mediate binding to negatively charged sulfate moieties on glycosaminoglycans

(GAGs)23. While mechanism for CFP binding to DNA is currently unknown, like GAG, DNA is a

Page 93: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

79

large polyanionic compound. The properdin residues proposed to bind sulfate moieties of GAGs may

also be responsible for mediating properdin deposition onto NETs23. However, as the mechanisms for

NETosis are still not fully understood, we cannot the rule out the process of mechanical mixing in the

neutrophil as granular proteins rupture and the nuclear envelope decondenses prior the release of

NETs.

5.3 Neutrophil Extracellular Traps Activate Complement

CFP deposition onto NETs implied that NETs may amplify complement AP through CFP

dependent mechanisms. In addition, NETs are void of CFH, therefore they act as an activating

surface because they do not contain the proteins necessary for regulating complement AP203

. We

were able to successfully show the activation of complement on NETs through spinning disc

immunofluorescence confocal microscopy. Looking downstream to the final component of

activation, we were able to detect the formation of C5b-9 in the presence of NETs (Figure 15).

This process was dependent on NETs as activation of neutrophils in the presence of DNase I

completely abolished C5b-9 formation. To our knowledge, this is the first time anyone has shown a

direct link between NETs and complement activation leading to C5b-9. These data suggest that NETs

and complement interactions play a significant role in contributing to tissue damage during a

deleterious inflammatory response.

Mechanistically, we determined that complement activation on NETs is achieved through

multiple pathways. CFP is necessary for activation of complement AP on NETs as inhibition of CFP

is sufficient to stop the assembly of C5b-9 in 20% (v/v) plasma in AP buffer (Figure 17). The

inhibition of CFP in 20% (v/v) plasma in RPMI buffer was not sufficient in stopping the assembly of

C5b-9. Without chelation of Ca2+ or Mg2+ in RPMI buffer, all potential modes of complement

activation can be initiated. No current studies have demonstrated that MBL binding to NETs can

initiate complement LP, however the deposition of C1q on NETs has been shown to activate

complement CP157. In addition, neutrophil MPO and elastase have been associated with complement

activation. MPO production of HOCl oxidizes C5 and activates it to form C5b-9197. Neutrophil

elastase cleaves C5 to produce a functionally active C5b6 which can initiate the assembly of C5b-941.

Therefore, it may be possible for NETs to activate complement through non canonical pathways.

Page 94: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

80

5.4 Neutrophils form NETs in Response to Bacteria

Much of the previous work was conducted through activation with PMA. While these data

demonstrate clearly, the amplification of complement through NETs, little is known with regards

to how this interaction contributes to antimicrobial function. Using the SYTOX green plate

reader assay, we probed for the ability of neutrophils to form NETs in response to infection with

Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, and Bacillus subtilis

Consistent with literature120

, we were able to demonstrate that P. aeruginosa (Figure 18), and S.

aureus (Figure 19) were able to induce the formation of NETs. We also report that E. coli and B.

subtilis are poor inducers of NETosis. Activation of neutrophils at a multiplicity of 10 was

insufficient to induce NETosis, only activation of neutrophils with E. coli and B. subtilis at MOI

100 was able to induce NETosis (Figure 19).

We postulate that this may be indicative of the virulence of the pathogenic bacteria.

Pseudomonas aeruginosa and Staphyloccocus aureus are both opportunistic pathogens. P.

aeruginosa and S. aureus are opportunistic pathogens that often establish themselves in

immunocompromised individuals (e.g. patients with cystic fibrosis)222

. P. aeruginosa is a major

cause of infections worldwide (10% of all infections in EU hospitals) and is a series threat to

public health223

. S. aureus is a common cause for sepsis with a significant associated rate of

mortality224

. Neutrophils may respond to these virulent strains of bacteria with an “exaggerated”

response by forming NETs. Whether the formation of NETs in response to virulent strains of

bacteria attenuates or exacerbates disease prognosis has yet to be determined. In contrast, E. coli

and B. subtilis are considered non pathogenic bacteria which are often found in the human

gastrointenstinal tract. However, virulent strains of these bacteria do exist. E. coli O157:H7, for

example, is a virulent strain that causes hemolytic uremic syndrome (HUS), a disease

characterized by bloody diarrhea, microangiopathic hemolytic anemia, thrombocytopenia, and

renal impairment53

. Currently, no research has been conducted with regards to neutrophil

induction of NETosis with E. coli O157:H7. In the future, it may be worthy to investigate

whether virulent strains of different strains of bacteria are able to induce the formation of NETs.

5.5 NETs Formed in Response to Bacteria Activate Complement

To further explore the interaction between complement and NETs, and understanding how they

may facilitate an antimicrobial response, we looked at the interaction between NETS and P.

Page 95: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

81

aeruginosa. We chose to focus on gram-negative P. aeruginosa because the pattern recognition

molecule properties of CFP have been demonstrated to be able to bind bacterial LPS30

. To study

NET, CFP, P. aeruginosa interactions, we utilized a strain of P. aeruginosa PAKgfp that

expresses green fluorescent protein (GFP) through a constitutively active promoter (a gift from

the lab of Dr. Marina Ulanova, Northern Ontario School of Medicine)216

. While we were able

demonstrate the deposition of CFP onto pseudomonas (Figure 20), our data indicate that this

phenomenon occurs independent of NETs. Thus, neutrophil CFP found on NETs is not actively

targeted to bacterial surfaces to mount an antimicrobial response.

While NETs do not actively deposit CFP onto bacteria, our data clearly illustrate the

bacterial trapping capabilities of NETs (Figure 20). PAKgfp trapped in NETs (Figure 20BEHK)

can be found at a much higher concentration compared to neutrophils that are activated with

PAKgfp in the presence of DNase I to disseminate NETs (Figure 20CFIL). We previously

demonstrate that NETs, formed in response to PMA, can act as an activating surface to propagate

the formation of C5b-9 (Figure 15). Although, NETs do not actively target CFP onto bacteria,

bacterial trapping and complement amplification can work together to mount an effective

immune response. We then asked whether NETs formed in response to infection can initiate a

complement response. Our data demonstrate that NETs formed in response to PAKgfp can

initiate complement activation leading to the formation of C5b-9 (Figure 21). This process was

dependent on utilizing NETs as a complement activating surface, because activating neutrophils

in the presence of DNase I was able to abolish the formation of complement C5b-9. To our

knowledge, this is the first demonstration linking NETs to bacterial trapping and complement

amplification. These data suggest that in response to infection, neutrophils may form NETS to

facilitate an effective immune response. However, this also suggests that if this process

overwhelms regulatory mechanisms, infections can trigger an unwanted deleterious

inflammatory pathology.

Mechanistically, NETs that are formed in response to P. aeruginosa activate complement

in the same manner as NETs formed in response to PMA. The activation of complement AP on

NETs formed in response to bacteria is dependent on CFP as inhibition of CFP with mAb is

sufficient to stop the assembly of C5b-9 in 20% (v/v) plasma in AP buffer (Figure 23B). The

addition of mAb to CFP to 20% (v/v) plasma in RPMI was not sufficient in stopping the formation of

C5b-9 on NETs formed in response to bacteria (Figure 23D). Thus, in addition to complement AP,

Page 96: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

82

other pathways of complement activation are at work on NETs. Therefore, in addition to containing

many antimicrobial peptides, the activation of complement on NETs may be an effective method for

antimicrobial clearance.

Integrating our findings from the interaction between complement and neutrophils, we

propose a model where infectious triggers can initiate a neutrophil response for mobilization and

activation (Figure 24). Neutrophil activation prompts the secretion of complement proteins contained

in neutrophils to modulate and activate the complement response. In addition, the formation of NETs

is effective in capturing bacteria and initiates complement amplification. Overall, the interactions

between complement and neutrophils may work together to mount an effective immune response.

Page 97: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

83

Figure 24. Proposed model of complement-neutrophil interactions. The activation of neutrophils leads to the secretion of complement proteins. Secretion of CFP is

capable of binding to invading microbes. Further activation of neutrophils will lead to the

formation of NETs. CFP is able to bind to both bacteria and NETs, however whether CFP

enhances the ability for NETs to capture bacteria has yet to be determined. NETs initiate the

amplification of multiple complement pathways which leads to the formation of C5b-9. The

activation of complement AP initiated by NETs is dependent on CFP.

Page 98: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

84

6 Conclusion and Future Directions

6.1 Conclusions

From the work presented here, we conclude that neutrophils contain key proteins of the

complement alternative pathway. This further strengthens the notion that neutrophils are key

mediators of inflammatory and immune responses. Neutrophil activation coordinates a strict

regiment to deal with potential threats (e.g. infection or tissue damage) through programmed

phagocytosis, degranulation and NETosis. We show that in response to activating stimuli (e.g.

C5a, fMLP, and PMA) complement proteins contained in neutrophils are secreted. Also, secreted

properdin is effective in recognizing and binding to P. aeruginosa. In addition, neutrophil

properdin was demonstrated to be deposited on NETs. The addition of complement competent

plasma to NETs revealed that they are able to initiate and activate complement. Inhibition of

properdin in AP buffer was sufficient in stopping the formation of C5b-9, however it could not

stop the formation of C5b-9 in RPMI buffer. Thus, we demonstrate that while NETs are able to

initiate complement AP, they also activate complement through other initiating pathways.

Neutrophils are a major determinant of local complement AP activity as neutrophil

release of CFP is able to dramatically increase local concentrations64

. Therefore we proposed that

NETs may also lead to activation of complement AP. We successfully demonstrated that NETs

can activate complement, however, a caveat to our current finding is that we were not be able to

demonstrate that this property is dependent on neutrophil properdin. Circulating CFP is found at

relatively low concentrations in plasma (4-6 μg/ml), however this concentration is still sufficient

in stabilizing the complement AP C3 convertase to amplify this constitutively active pathway63

.

Further research needs to be conducted to elucidate the mechanisms by which NETs activate

complement. In addition, while our ex vivo model successfully demonstrates that NETs are able

to activate complement, further research needs to be conducted to investigate whether this

interaction can contribute to anti microbial activity or endothelial cell damage.

6.2 Future Directions

Our work demonstrates the potential for NETs to interact with complement to mediate an

inflammatory response. There are still many questions which remain to be addressed to bridge

Page 99: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

85

the gap between this ex vivo model and understanding how this interaction can contribute to

microbial clearance or pathology in an in vivo setting.

We demonstrated that NETs are formed in response to PMA, P. aeruginosa, and S.

aureus. The activation of neutrophils with C5a did not induce the formation of NETs, however, a

study has demonstrated that C5a primed neutrophils enhances neutrophil oxidative burst in

response to ANCA220

. Since the formation of NETs is largely dependent on production of ROS,

it is important to address whether C5a primed neutrophils are more susceptible to NETosis, thus

further bridging the gap between complement and neutrophils.

Our analysis using spinning-disc confocal microscopy reveals that CFP is found on

NETs, however we were unable to identify the presence of the other complement proteins

contained in neutrophils on the NETs. As we alluded to earlier, at this time, we can not conclude

that these proteins are not found in NETs as the low concentration of these proteins may be

below the detection limits for immunofluorescence microscopy. In order to address this question,

an optimized technique for excising proteins from NETs needs to be established and more

sensitive methods (e.g. ELISA or Mass-spectrometry) need to be utilized to definitively answer

this question.

In our hypothesis, we proposed that neutrophils mount a targeted antimicrobial response

through NETs to initiate complement. We anticipated that NETs would enhance the deposition

of CFP onto bacteria. Our finding that NETs do not enhance the deposition of CFP onto P.

aeruginosa was an interesting revelation. However, the ability of CFP to act as a pattern

recognition molecule allows it to bind various ligands. In addition, CFP exists as a multimer

allowing it to interact with multiple ligands. CFP has been shown to bind to both LPS and DNA.

Therefore, it is likely that the deposition of CFP onto P. aeruginosa may enhance the ability of

NETs to trap bacteria. Further studies need to be conducted to address this.

In order to look for the activation of complement on NETs, we probed for the assembly

of C5b-9. Our data clearly demonstrate that NETs can activate complement, and that

complement AP activation on NETs is dependent on CFP. However, the formation of C5b-9 on

NETs is somewhat puzzling. At this moment, there does not appear to be a discernible pattern for

C5b-9 deposition. C5b-9 does not appear to show preference for NET DNA, nor does it show

preference to PAKgfp. However, C5b-9 does appear to be localized to various clusters scattered

Page 100: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

86

throughout the NET. This suggests that there is an activating factor which directs the formation

of C5b-9 on NETs. Further studies need to be conducted to identify where C5b-9 is being

assembled on NETs.

We also need to address the caveat in our current finding. While we show that NETs are

complement activating, we have not shown that this process is dependent on neutrophil CFP. To

fully understand the mechanism on how NETs can activate the different complement initiating

pathways, we need to utilize the full arsenal of inhibitory antibodies against complement proteins

and utilize complement-depleted plasma, which is commercially available . However, addressing

this question will be technically challenging, because complement depleted plasmas are often

also complement in active. We may also utilize properdin knockout mouse models to address

this caveat, however the murine complement system is different from the human complement

system. Complement deficient mice do not display the same pathological symptoms or

phenotypes as their human counterparts.

Our ex vivo model successfully demonstrated that NETs are able to activate complement

activity. However, whether this process mounts an antimicrobial response or contributes to cell

damage remains to be addressed. To address whether complement and NETs work together to

mount an antimicrobial response, bacteria colony counts need to be performed in the presence or

absence of NETs and/or complement competent/inactive plasma. The introduction of endothelial

cells to our ex vivo model (e.g. HUVEC, BOEC)225

will be required to address whether

complement-NET interactions can lead to endothelial cell damage.

Neutrophils have often been shown to play an important, but poorly understood role in

complement mediated pathologies. Here we show that NETs are able to initiate complement

activity. However, in patients who have deficiencies in complement regulatory proteins, this

localized activation of complement may overwhelm the delicate balance of complement

activation and regulation to precipitate into pathology. It would be worth investigating if there is

increased complement activity on NETs for patients who have known mutations in complement

regulatory proteins. This work may answer many of the mysteries behind complement mutations

and pathology.

Page 101: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

87

7 References

1. Sunyer, J.O., Zarkadis, I.K. & Lambris, J.D. Complement diversity: a mechanism for

generating immune diversity? Immunol Today 19, 519-523 (1998).

2. Ricklin, D., Hajishengallis, G., Yang, K. & Lambris, J.D. Complement: a key system for

immune surveillance and homeostasis. Nat Immunol 11, 785-797 (2010).

3. Sjoberg, A.P., Trouw, L.A. & Blom, A.M. Complement activation and inhibition: a

delicate balance. Trends Immunol 30, 83-90 (2009).

4. Markiewski, M.M. & Lambris, J.D. The role of complement in inflammatory diseases

from behind the scenes into the spotlight. Am J Pathol 171, 715-727 (2007).

5. Walport, M.J. Complement. First of two parts. N Engl J Med 344, 1058-1066 (2001).

6. Walport, M.J. Complement. Second of two parts. N Engl J Med 344, 1140-1144 (2001).

7. Huber-Lang, M., et al. Generation of C5a in the absence of C3: a new complement

activation pathway. Nat Med 12, 682-687 (2006).

8. Gal, P., Barna, L., Kocsis, A. & Zavodszky, P. Serine proteases of the classical and lectin

pathways: similarities and differences. Immunobiology 212, 267-277 (2007).

9. Wallis, R., Mitchell, D.A., Schmid, R., Schwaeble, W.J. & Keeble, A.H. Paths reunited:

Initiation of the classical and lectin pathways of complement activation. Immunobiology

215, 1-11 (2010).

10. Gaboriaud, C., et al. Structure and activation of the C1 complex of complement:

unraveling the puzzle. Trends Immunol 25, 368-373 (2004).

11. Arlaud, G.J., et al. Structural biology of the C1 complex of complement unveils the

mechanisms of its activation and proteolytic activity. Mol Immunol 39, 383-394 (2002).

12. Holmskov, U., Malhotra, R., Sim, R.B. & Jensenius, J.C. Collectins: collagenous C-type

lectins of the innate immune defense system. Immunol Today 15, 67-74 (1994).

13. Weis, W.I., Drickamer, K. & Hendrickson, W.A. Structure of a C-type mannose-binding

protein complexed with an oligosaccharide. Nature 360, 127-134 (1992).

14. Drickamer, K. Engineering galactose-binding activity into a C-type mannose-binding

protein. Nature 360, 183-186 (1992).

15. Sato, T., Endo, Y., Matsushita, M. & Fujita, T. Molecular characterization of a novel

serine protease involved in activation of the complement system by mannose-binding

protein. Int Immunol 6, 665-669 (1994).

16. Thiel, S., et al. A second serine protease associated with mannan-binding lectin that

activates complement. Nature 386, 506-510 (1997).

Page 102: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

88

17. Gadjeva, M., Thiel, S. & Jensenius, J.C. The mannan-binding-lectin pathway of the

innate immune response. Curr Opin Immunol 13, 74-78 (2001).

18. Chen, C.B. & Wallis, R. Two mechanisms for mannose-binding protein modulation of

the activity of its associated serine proteases. J Biol Chem 279, 26058-26065 (2004).

19. Sahu, A., Kozel, T.R. & Pangburn, M.K. Specificity of the thioester-containing reactive

site of human C3 and its significance to complement activation. Biochem J 302 ( Pt 2),

429-436 (1994).

20. Pangburn, M.K., Schreiber, R.D. & Muller-Eberhard, H.J. Formation of the initial C3

convertase of the alternative complement pathway. Acquisition of C3b-like activities by

spontaneous hydrolysis of the putative thioester in native C3. J Exp Med 154, 856-867

(1981).

21. Pangburn, M.K. & Muller-Eberhard, H.J. The C3 convertase of the alternative pathway

of human complement. Enzymic properties of the bimolecular proteinase. Biochem J 235,

723-730 (1986).

22. Medicus, R.G., Gotze, O. & Muller-Eberhard, H.J. Alternative pathway of complement:

recruitment of precursor properdin by the labile C3/C5 convertase and the potentiation of

the pathway. J Exp Med 144, 1076-1093 (1976).

23. Kemper, C., Atkinson, J.P. & Hourcade, D.E. Properdin: emerging roles of a pattern-

recognition molecule. Annu Rev Immunol 28, 131-155 (2010).

24. Zipfel, P.F., et al. Factor H family proteins: on complement, microbes and human

diseases. Biochem Soc Trans 30, 971-978 (2002).

25. Weiler, J.M. Regulation of C5 convertase activity by properdin, factors B and H.

Immunol Res 8, 305-315 (1989).

26. Weiler, J.M., Daha, M.R., Austen, K.F. & Fearon, D.T. Control of the amplification

convertase of complement by the plasma protein beta1H. Proc Natl Acad Sci U S A 73,

3268-3272 (1976).

27. Farries, T.C., Lachmann, P.J. & Harrison, R.A. Analysis of the interactions between

properdin, the third component of complement (C3), and its physiological activation

products. Biochem J 252, 47-54 (1988).

28. Spitzer, D., Mitchell, L.M., Atkinson, J.P. & Hourcade, D.E. Properdin can initiate

complement activation by binding specific target surfaces and providing a platform for de

novo convertase assembly. J Immunol 179, 2600-2608 (2007).

29. Kemper, C., Mitchell, L.M., Zhang, L. & Hourcade, D.E. The complement protein

properdin binds apoptotic T cells and promotes complement activation and phagocytosis.

Proc Natl Acad Sci U S A 105, 9023-9028 (2008).

Page 103: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

89

30. Kimura, Y., Miwa, T., Zhou, L. & Song, W.C. Activator-specific requirement of

properdin in the initiation and amplification of the alternative pathway complement.

Blood 111, 732-740 (2008).

31. Xu, W., et al. Properdin binds to late apoptotic and necrotic cells independently of C3b

and regulates alternative pathway complement activation. J Immunol 180, 7613-7621

(2008).

32. Zhang, L., Lawrence, R., Frazier, B.A. & Esko, J.D. CHO glycosylation mutants:

proteoglycans. Methods Enzymol 416, 205-221 (2006).

33. Nolan, K.F., Schwaeble, W., Kaluz, S., Dierich, M.P. & Reid, K.B. Molecular cloning of

the cDNA coding for properdin, a positive regulator of the alternative pathway of human

complement. Eur J Immunol 21, 771-776 (1991).

34. Pangburn, M.K. Analysis of the natural polymeric forms of human properdin and their

functions in complement activation. J Immunol 142, 202-207 (1989).

35. Sun, Z., Reid, K.B. & Perkins, S.J. The dimeric and trimeric solution structures of the

multidomain complement protein properdin by X-ray scattering, analytical

ultracentrifugation and constrained modelling. J Mol Biol 343, 1327-1343 (2004).

36. Smith, C.A., Pangburn, M.K., Vogel, C.W. & Muller-Eberhard, H.J. Molecular

architecture of human properdin, a positive regulator of the alternative pathway of

complement. J Biol Chem 259, 4582-4588 (1984).

37. Tan, K., et al. Crystal structure of the TSP-1 type 1 repeats: a novel layered fold and its

biological implication. J Cell Biol 159, 373-382 (2002).

38. Harboe, M. & Mollnes, T.E. The alternative complement pathway revisited. J Cell Mol

Med 12, 1074-1084 (2008).

39. Pangburn, M.K. & Rawal, N. Structure and function of complement C5 convertase

enzymes. Biochem Soc Trans 30, 1006-1010 (2002).

40. Muller-Eberhard, H.J. The membrane attack complex of complement. Annu Rev Immunol

4, 503-528 (1986).

41. Vogt, W. Cleavage of the fifth component of complement and generation of a

functionally active C5b6-like complex by human leukocyte elastase. Immunobiology 201,

470-477 (2000).

42. Kohl, J. Self, non-self, and danger: a complementary view. Adv Exp Med Biol 586, 71-94

(2006).

43. Nathan, C. Points of control in inflammation. Nature 420, 846-852 (2002).

44. Levi, M. & van der Poll, T. Inflammation and coagulation. Crit Care Med 38, S26-34

(2010).

Page 104: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

90

45. Ricklin, D. & Lambris, J.D. Complement in immune and inflammatory disorders:

pathophysiological mechanisms. J Immunol 190, 3831-3838 (2013).

46. Carroll, M.C. The complement system in regulation of adaptive immunity. Nat Immunol

5, 981-986 (2004).

47. Mastellos, D. & Lambris, J.D. Cross-disciplinary research stirs new challenges into the

study of the structure, function and systems biology of complement. Adv Exp Med Biol

586, 1-16 (2006).

48. Thurman, J.M. & Holers, V.M. The central role of the alternative complement pathway in

human disease. J Immunol 176, 1305-1310 (2006).

49. Holers, V.M. The complement system as a therapeutic target in autoimmunity. Clin

Immunol 107, 140-151 (2003).

50. Mizuno, M., et al. Membrane complement regulators protect against the development of

type II collagen-induced arthritis in rats. Arthritis Rheum 44, 2425-2434 (2001).

51. Ratnoff, W.D., Fearon, D.T. & Austen, K.F. The role of antibody in the activation of the

alternative complement pathway. Springer Semin Immunopathol 6, 361-371 (1983).

52. Wang, Y., et al. Amelioration of lupus-like autoimmune disease in NZB/WF1 mice after

treatment with a blocking monoclonal antibody specific for complement component C5.

Proc Natl Acad Sci U S A 93, 8563-8568 (1996).

53. Noris, M. & Remuzzi, G. Hemolytic uremic syndrome. J Am Soc Nephrol 16, 1035-1050

(2005).

54. Thorpe, C.M. Shiga toxin-producing Escherichia coli infection. Clin Infect Dis 38, 1298-

1303 (2004).

55. Karmali, M.A., Steele, B.T., Petric, M. & Lim, C. Sporadic cases of haemolytic-uraemic

syndrome associated with faecal cytotoxin and cytotoxin-producing Escherichia coli in

stools. Lancet 1, 619-620 (1983).

56. Riley, L.W., et al. Hemorrhagic colitis associated with a rare Escherichia coli serotype. N

Engl J Med 308, 681-685 (1983).

57. Varma, J.K., et al. An outbreak of Escherichia coli O157 infection following exposure to

a contaminated building. JAMA 290, 2709-2712 (2003).

58. Obrig, T.G., Moran, T.P. & Brown, J.E. The mode of action of Shiga toxin on peptide

elongation of eukaryotic protein synthesis. Biochem J 244, 287-294 (1987).

59. Erwert, R.D., et al. Shiga toxin induces decreased expression of the anti-apoptotic protein

Mcl-1 concomitant with the onset of endothelial apoptosis. Microb Pathog 35, 87-93

(2003).

Page 105: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

91

60. Orth, D., et al. Shiga toxin activates complement and binds factor H: evidence for an

active role of complement in hemolytic uremic syndrome. J Immunol 182, 6394-6400

(2009).

61. Thurman, J.M., et al. Alternative pathway of complement in children with diarrhea-

associated hemolytic uremic syndrome. Clin J Am Soc Nephrol 4, 1920-1924 (2009).

62. Lapeyraque, A.L., et al. Eculizumab in severe Shiga-toxin-associated HUS. N Engl J

Med 364, 2561-2563 (2011).

63. Nolan, K.F. & Reid, K.B. Properdin. Methods Enzymol 223, 35-46 (1993).

64. Schwaeble, W.J. & Reid, K.B. Does properdin crosslink the cellular and the humoral

immune response? Immunol Today 20, 17-21 (1999).

65. Forsyth, K.D., Simpson, A.C., Fitzpatrick, M.M., Barratt, T.M. & Levinsky, R.J.

Neutrophil-mediated endothelial injury in haemolytic uraemic syndrome. Lancet 2, 411-

414 (1989).

66. Roumenina, L.T., et al. A prevalent C3 mutation in aHUS patients causes a direct C3

convertase gain of function. Blood 119, 4182-4191 (2012).

67. Sartz, L., et al. A novel C3 mutation causing increased formation of the C3 convertase in

familial atypical hemolytic uremic syndrome. J Immunol 188, 2030-2037 (2012).

68. Lhotta, K., et al. A large family with a gain-of-function mutation of complement C3

predisposing to atypical hemolytic uremic syndrome, microhematuria, hypertension and

chronic renal failure. Clin J Am Soc Nephrol 4, 1356-1362 (2009).

69. Goicoechea de Jorge, E., et al. Gain-of-function mutations in complement factor B are

associated with atypical hemolytic uremic syndrome. Proc Natl Acad Sci U S A 104, 240-

245 (2007).

70. Tawadrous, H., et al. A novel mutation in the complement factor B gene (CFB) and

atypical hemolytic uremic syndrome. Pediatr Nephrol 25, 947-951 (2010).

71. Warwicker, P., et al. Genetic studies into inherited and sporadic hemolytic uremic

syndrome. Kidney Int 53, 836-844 (1998).

72. Ying, L., et al. Complement factor H gene mutation associated with autosomal recessive

atypical hemolytic uremic syndrome. Am J Hum Genet 65, 1538-1546 (1999).

73. Dragon-Durey, M.A., et al. Heterozygous and homozygous factor h deficiencies

associated with hemolytic uremic syndrome or membranoproliferative

glomerulonephritis: report and genetic analysis of 16 cases. J Am Soc Nephrol 15, 787-

795 (2004).

74. Jozsi, M., et al. Factor H autoantibodies in atypical hemolytic uremic syndrome correlate

with CFHR1/CFHR3 deficiency. Blood 111, 1512-1514 (2008).

Page 106: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

92

75. Dragon-Durey, M.A., et al. Anti-Factor H autoantibodies associated with atypical

hemolytic uremic syndrome. J Am Soc Nephrol 16, 555-563 (2005).

76. Noris, M. & Remuzzi, G. Atypical hemolytic-uremic syndrome. N Engl J Med 361,

1676-1687 (2009).

77. Allen, U. & Licht, C. Pandemic H1N1 influenza A infection and (atypical) HUS--more

than just another trigger? Pediatr Nephrol 26, 3-5 (2011).

78. Printza, N., et al. Pandemic influenza A (H1N1) 2009-associated hemolytic uremic

syndrome. Pediatr Nephrol 26, 143-144 (2011).

79. Golubovic, E., Miljkovic, P., Zivic, S., Jovancic, D. & Kostic, G. Hemolytic uremic

syndrome associated with novel influenza A H1N1 infection. Pediatr Nephrol 26, 149-

150 (2011).

80. Trachtman, H., et al. Atypical hemolytic uremic syndrome associated with H1N1

influenza A virus infection. Pediatr Nephrol 26, 145-146 (2011).

81. Caprioli, J., et al. Genetics of HUS: the impact of MCP, CFH, and IF mutations on

clinical presentation, response to treatment, and outcome. Blood 108, 1267-1279 (2006).

82. Kavanagh, D., Goodship, T.H. & Richards, A. Atypical haemolytic uraemic syndrome.

Br Med Bull 77-78, 5-22 (2006).

83. Girardi, G., et al. Complement C5a receptors and neutrophils mediate fetal injury in the

antiphospholipid syndrome. J Clin Invest 112, 1644-1654 (2003).

84. Nathan, C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6,

173-182 (2006).

85. Lieschke, G.J., et al. Mice lacking granulocyte colony-stimulating factor have chronic

neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired

neutrophil mobilization. Blood 84, 1737-1746 (1994).

86. Le Cabec, V., Cowland, J.B., Calafat, J. & Borregaard, N. Targeting of proteins to

granule subsets is determined by timing and not by sorting: The specific granule protein

NGAL is localized to azurophil granules when expressed in HL-60 cells. Proc Natl Acad

Sci U S A 93, 6454-6457 (1996).

87. Borregaard, N., Sorensen, O.E. & Theilgaard-Monch, K. Neutrophil granules: a library of

innate immunity proteins. Trends Immunol 28, 340-345 (2007).

88. Faurschou, M., Sorensen, O.E., Johnsen, A.H., Askaa, J. & Borregaard, N. Defensin-rich

granules of human neutrophils: characterization of secretory properties. Biochim Biophys

Acta 1591, 29-35 (2002).

89. Amulic, B., Cazalet, C., Hayes, G.L., Metzler, K.D. & Zychlinsky, A. Neutrophil

function: from mechanisms to disease. Annu Rev Immunol 30, 459-489 (2012).

Page 107: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

93

90. Rorvig, S., et al. Ficolin-1 is present in a highly mobilizable subset of human neutrophil

granules and associates with the cell surface after stimulation with fMLP. J Leukoc Biol

86, 1439-1449 (2009).

91. Theilgaard-Monch, K., et al. The transcriptional program of terminal granulocytic

differentiation. Blood 105, 1785-1796 (2005).

92. Borregaard, N., et al. Stimulus-dependent secretion of plasma proteins from human

neutrophils. J Clin Invest 90, 86-96 (1992).

93. Mantovani, A., Cassatella, M.A., Costantini, C. & Jaillon, S. Neutrophils in the activation

and regulation of innate and adaptive immunity. Nat Rev Immunol 11, 519-531 (2011).

94. Lawrence, M.B., Kansas, G.S., Kunkel, E.J. & Ley, K. Threshold levels of fluid shear

promote leukocyte adhesion through selectins (CD62L,P,E). J Cell Biol 136, 717-727

(1997).

95. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 33, 657-670 (2010).

96. McEver, R.P. & Cummings, R.D. Role of PSGL-1 binding to selectins in leukocyte

recruitment. J Clin Invest 100, S97-103 (1997).

97. Ley, K., Laudanna, C., Cybulsky, M.I. & Nourshargh, S. Getting to the site of

inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 7, 678-689

(2007).

98. McDonald, B., et al. Intravascular danger signals guide neutrophils to sites of sterile

inflammation. Science 330, 362-366 (2010).

99. Zarbock, A. & Ley, K. Mechanisms and consequences of neutrophil interaction with the

endothelium. Am J Pathol 172, 1-7 (2008).

100. Brinkmann, V., et al. Neutrophil extracellular traps kill bacteria. Science 303, 1532-1535

(2004).

101. Henson, P.M. & Johnston, R.B., Jr. Tissue injury in inflammation. Oxidants, proteinases,

and cationic proteins. J Clin Invest 79, 669-674 (1987).

102. Weiss, S.J. Tissue destruction by neutrophils. N Engl J Med 320, 365-376 (1989).

103. Stark, M.A., et al. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-

23 and IL-17. Immunity 22, 285-294 (2005).

104. Serhan, C.N., Yacoubian, S. & Yang, R. Anti-inflammatory and proresolving lipid

mediators. Annu Rev Pathol 3, 279-312 (2008).

105. Schwab, J.M., Chiang, N., Arita, M. & Serhan, C.N. Resolvin E1 and protectin D1

activate inflammation-resolution programmes. Nature 447, 869-874 (2007).

Page 108: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

94

106. Oh, S.F., Pillai, P.S., Recchiuti, A., Yang, R. & Serhan, C.N. Pro-resolving actions and

stereoselective biosynthesis of 18S E-series resolvins in human leukocytes and murine

inflammation. J Clin Invest 121, 569-581 (2011).

107. Fuchs, T.A., et al. Novel cell death program leads to neutrophil extracellular traps. J Cell

Biol 176, 231-241 (2007).

108. Papayannopoulos, V. & Zychlinsky, A. NETs: a new strategy for using old weapons.

Trends Immunol 30, 513-521 (2009).

109. Urban, C.F., et al. Neutrophil extracellular traps contain calprotectin, a cytosolic protein

complex involved in host defense against Candida albicans. PLoS Pathog 5, e1000639

(2009).

110. Wang, Y., et al. Human PAD4 regulates histone arginine methylation levels via

demethylimination. Science 306, 279-283 (2004).

111. Li, P., et al. PAD4 is essential for antibacterial innate immunity mediated by neutrophil

extracellular traps. J Exp Med 207, 1853-1862 (2010).

112. Papayannopoulos, V., Metzler, K.D., Hakkim, A. & Zychlinsky, A. Neutrophil elastase

and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol

191, 677-691 (2010).

113. Bianchi, M., et al. Restoration of NET formation by gene therapy in CGD controls

aspergillosis. Blood 114, 2619-2622 (2009).

114. Metzler, K.D., et al. Myeloperoxidase is required for neutrophil extracellular trap

formation: implications for innate immunity. Blood 117, 953-959 (2011).

115. Yost, C.C., et al. Impaired neutrophil extracellular trap (NET) formation: a novel innate

immune deficiency of human neonates. Blood 113, 6419-6427 (2009).

116. Hakkim, A., et al. Activation of the Raf-MEK-ERK pathway is required for neutrophil

extracellular trap formation. Nat Chem Biol 7, 75-77 (2011).

117. Remijsen, Q., et al. Neutrophil extracellular trap cell death requires both autophagy and

superoxide generation. Cell Res 21, 290-304 (2011).

118. Clark, S.R., et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare

bacteria in septic blood. Nat Med 13, 463-469 (2007).

119. Tadie, J.M., et al. HMGB1 promotes neutrophil extracellular trap formation through

interactions with Toll-like receptor 4. Am J Physiol Lung Cell Mol Physiol 304, L342-

349 (2013).

120. Cheng, O.Z. & Palaniyar, N. NET balancing: a problem in inflammatory lung diseases.

Front Immunol 4, 1 (2013).

Page 109: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

95

121. Pilsczek, F.H., et al. A novel mechanism of rapid nuclear neutrophil extracellular trap

formation in response to Staphylococcus aureus. J Immunol 185, 7413-7425 (2010).

122. Grinberg, N., Elazar, S., Rosenshine, I. & Shpigel, N.Y. Beta-hydroxybutyrate abrogates

formation of bovine neutrophil extracellular traps and bactericidal activity against

mammary pathogenic Escherichia coli. Infect Immun 76, 2802-2807 (2008).

123. Young, R.L., et al. Neutrophil extracellular trap (NET)-mediated killing of Pseudomonas

aeruginosa: evidence of acquired resistance within the CF airway, independent of CFTR.

PLoS One 6, e23637 (2011).

124. Douda, D.N., Jackson, R., Grasemann, H. & Palaniyar, N. Innate immune collectin

surfactant protein D simultaneously binds both neutrophil extracellular traps and

carbohydrate ligands and promotes bacterial trapping. J Immunol 187, 1856-1865 (2011).

125. Buchanan, J.T., et al. DNase expression allows the pathogen group A Streptococcus to

escape killing in neutrophil extracellular traps. Curr Biol 16, 396-400 (2006).

126. Crotty Alexander, L.E., et al. M1T1 group A streptococcal pili promote epithelial

colonization but diminish systemic virulence through neutrophil extracellular entrapment.

J Mol Med (Berl) 88, 371-381 (2010).

127. Beiter, K., et al. An endonuclease allows Streptococcus pneumoniae to escape from

neutrophil extracellular traps. Curr Biol 16, 401-407 (2006).

128. Ramos-Kichik, V., et al. Neutrophil extracellular traps are induced by Mycobacterium

tuberculosis. Tuberculosis (Edinb) 89, 29-37 (2009).

129. Guimaraes-Costa, A.B., et al. Leishmania amazonensis promastigotes induce and are

killed by neutrophil extracellular traps. Proc Natl Acad Sci U S A 106, 6748-6753 (2009).

130. Gabriel, C., McMaster, W.R., Girard, D. & Descoteaux, A. Leishmania donovani

promastigotes evade the antimicrobial activity of neutrophil extracellular traps. J

Immunol 185, 4319-4327 (2010).

131. Abi Abdallah, D.S., et al. Toxoplasma gondii triggers release of human and mouse

neutrophil extracellular traps. Infect Immun 80, 768-777 (2012).

132. Bruns, S., et al. Production of extracellular traps against Aspergillus fumigatus in vitro

and in infected lung tissue is dependent on invading neutrophils and influenced by

hydrophobin RodA. PLoS Pathog 6, e1000873 (2010).

133. Urban, C.F., Reichard, U., Brinkmann, V. & Zychlinsky, A. Neutrophil extracellular

traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol 8, 668-

676 (2006).

134. Saitoh, T., et al. Neutrophil extracellular traps mediate a host defense response to human

immunodeficiency virus-1. Cell Host Microbe 12, 109-116 (2012).

Page 110: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

96

135. Narasaraju, T., et al. Excessive neutrophils and neutrophil extracellular traps contribute

to acute lung injury of influenza pneumonitis. Am J Pathol 179, 199-210 (2011).

136. Ng, H.H., et al. Doxycycline treatment attenuates acute lung injury in mice infected with

virulent influenza H3N2 virus: involvement of matrix metalloproteinases. Exp Mol

Pathol 92, 287-295 (2012).

137. Yousefi, S., Mihalache, C., Kozlowski, E., Schmid, I. & Simon, H.U. Viable neutrophils

release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ 16,

1438-1444 (2009).

138. Gupta, A.K., Hasler, P., Holzgreve, W., Gebhardt, S. & Hahn, S. Induction of neutrophil

extracellular DNA lattices by placental microparticles and IL-8 and their presence in

preeclampsia. Hum Immunol 66, 1146-1154 (2005).

139. Nishinaka, Y., Arai, T., Adachi, S., Takaori-Kondo, A. & Yamashita, K. Singlet oxygen

is essential for neutrophil extracellular trap formation. Biochem Biophys Res Commun

413, 75-79 (2011).

140. Kraemer, B.F., et al. Novel anti-bacterial activities of beta-defensin 1 in human platelets:

suppression of pathogen growth and signaling of neutrophil extracellular trap formation.

PLoS Pathog 7, e1002355 (2011).

141. Neeli, I., Khan, S.N. & Radic, M. Histone deimination as a response to inflammatory

stimuli in neutrophils. J Immunol 180, 1895-1902 (2008).

142. Palic, D., Ostojic, J., Andreasen, C.B. & Roth, J.A. Fish cast NETs: neutrophil

extracellular traps are released from fish neutrophils. Dev Comp Immunol 31, 805-816

(2007).

143. Rada, B., et al. Pyocyanin-enhanced neutrophil extracellular trap formation requires the

NADPH oxidase. PLoS One 8, e54205 (2013).

144. Bratton, D.L. & Henson, P.M. Neutrophil clearance: when the party is over, clean-up

begins. Trends Immunol 32, 350-357 (2011).

145. Caielli, S., Banchereau, J. & Pascual, V. Neutrophils come of age in chronic

inflammation. Curr Opin Immunol 24, 671-677 (2012).

146. Kaplan, M.J. & Radic, M. Neutrophil extracellular traps: double-edged swords of innate

immunity. J Immunol 189, 2689-2695 (2012).

147. Xu, J., et al. Extracellular histones are major mediators of death in sepsis. Nat Med 15,

1318-1321 (2009).

148. Saffarzadeh, M., et al. Neutrophil extracellular traps directly induce epithelial and

endothelial cell death: a predominant role of histones. PLoS One 7, e32366 (2012).

Page 111: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

97

149. Gurevitz, S.L., Snyder, J.A., Wessel, E.K., Frey, J. & Williamson, B.A. Systemic lupus

erythematosus: a review of the disease and treatment options. Consult Pharm 28, 110-121

(2013).

150. Watanabe, H., et al. Modulation of renal disease in MRL/lpr mice genetically deficient in

the alternative complement pathway factor B. J Immunol 164, 786-794 (2000).

151. Elliott, M.K., et al. Effects of complement factor D deficiency on the renal disease of

MRL/lpr mice. Kidney Int 65, 129-138 (2004).

152. Bao, L., et al. Transgenic expression of a soluble complement inhibitor protects against

renal disease and promotes survival in MRL/lpr mice. J Immunol 168, 3601-3607 (2002).

153. Prodeus, A.P., et al. A critical role for complement in maintenance of self-tolerance.

Immunity 9, 721-731 (1998).

154. Liu, C.L., et al. Specific post-translational histone modifications of neutrophil

extracellular traps as immunogens and potential targets of lupus autoantibodies. Arthritis

Res Ther 14, R25 (2012).

155. Villanueva, E., et al. Netting neutrophils induce endothelial damage, infiltrate tissues,

and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol

187, 538-552 (2011).

156. Hakkim, A., et al. Impairment of neutrophil extracellular trap degradation is associated

with lupus nephritis. Proc Natl Acad Sci U S A 107, 9813-9818 (2010).

157. Leffler, J., et al. Neutrophil extracellular traps that are not degraded in systemic lupus

erythematosus activate complement exacerbating the disease. J Immunol 188, 3522-3531

(2012).

158. Majithia, V. & Geraci, S.A. Rheumatoid arthritis: diagnosis and management. Am J Med

120, 936-939 (2007).

159. Chang, M., et al. A large-scale rheumatoid arthritis genetic study identifies association at

chromosome 9q33.2. PLoS Genet 4, e1000107 (2008).

160. Ji, H., et al. Arthritis critically dependent on innate immune system players. Immunity 16,

157-168 (2002).

161. Hietala, M.A., Jonsson, I.M., Tarkowski, A., Kleinau, S. & Pekna, M. Complement

deficiency ameliorates collagen-induced arthritis in mice. J Immunol 169, 454-459

(2002).

162. Rohrbach, A.S., Hemmers, S., Arandjelovic, S., Corr, M. & Mowen, K.A. PAD4 is not

essential for disease in the K/BxN murine autoantibody-mediated model of arthritis.

Arthritis Res Ther 14, R104 (2012).

163. Pratesi, F., et al. Antibodies from patients with rheumatoid arthritis target citrullinated

histone 4 contained in neutrophils extracellular traps. Ann Rheum Dis (2013).

Page 112: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

98

164. Taube, C., Dakhama, A. & Gelfand, E.W. Insights into the pathogenesis of asthma

utilizing murine models. Int Arch Allergy Immunol 135, 173-186 (2004).

165. Bautsch, W., et al. Cutting edge: guinea pigs with a natural C3a-receptor defect exhibit

decreased bronchoconstriction in allergic airway disease: evidence for an involvement of

the C3a anaphylatoxin in the pathogenesis of asthma. J Immunol 165, 5401-5405 (2000).

166. Krug, N., Tschernig, T., Erpenbeck, V.J., Hohlfeld, J.M. & Kohl, J. Complement factors

C3a and C5a are increased in bronchoalveolar lavage fluid after segmental allergen

provocation in subjects with asthma. Am J Respir Crit Care Med 164, 1841-1843 (2001).

167. Hamelmann, E., et al. Development of eosinophilic airway inflammation and airway

hyperresponsiveness requires interleukin-5 but not immunoglobulin E or B lymphocytes.

Am J Respir Cell Mol Biol 21, 480-489 (1999).

168. Simpson, J.L., Scott, R., Boyle, M.J. & Gibson, P.G. Inflammatory subtypes in asthma:

assessment and identification using induced sputum. Respirology 11, 54-61 (2006).

169. Haldar, P. & Pavord, I.D. Noneosinophilic asthma: a distinct clinical and pathologic

phenotype. J Allergy Clin Immunol 119, 1043-1052; quiz 1053-1044 (2007).

170. Dworski, R., Simon, H.U., Hoskins, A. & Yousefi, S. Eosinophil and neutrophil

extracellular DNA traps in human allergic asthmatic airways. J Allergy Clin Immunol

127, 1260-1266 (2011).

171. Xiao, H., et al. Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase

cause glomerulonephritis and vasculitis in mice. J Clin Invest 110, 955-963 (2002).

172. Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J. & Jennette, J.C. Alternative complement

pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic

autoantibodies. Am J Pathol 170, 52-64 (2007).

173. Huugen, D., et al. Inhibition of complement factor C5 protects against anti-

myeloperoxidase antibody-mediated glomerulonephritis in mice. Kidney Int 71, 646-654

(2007).

174. Kallenberg, C.G., et al. ANCA--pathophysiology revisited. Clin Exp Immunol 100, 1-3

(1995).

175. Kessenbrock, K., et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat

Med 15, 623-625 (2009).

176. Welbourn, C.R. & Young, Y. Endotoxin, septic shock and acute lung injury: neutrophils,

macrophages and inflammatory mediators. Br J Surg 79, 998-1003 (1992).

177. Markiewski, M.M., DeAngelis, R.A. & Lambris, J.D. Complexity of complement

activation in sepsis. J Cell Mol Med 12, 2245-2254 (2008).

178. Mavrommatis, A.C., et al. Coagulation system and platelets are fully activated in

uncomplicated sepsis. Crit Care Med 28, 451-457 (2000).

Page 113: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

99

179. Fuchs, T.A., et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S

A 107, 15880-15885 (2010).

180. Semeraro, F., et al. Extracellular histones promote thrombin generation through platelet-

dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood 118, 1952-1961

(2011).

181. McDonald, B. & Kubes, P. Neutrophils and intravascular immunity in the liver during

infection and sterile inflammation. Toxicol Pathol 40, 157-165 (2012).

182. Nishiura, H. The alternative C5a receptor function. Adv Exp Med Biol 735, 111-121

(2013).

183. Martin, U., et al. The human C3a receptor is expressed on neutrophils and monocytes,

but not on B or T lymphocytes. J Exp Med 186, 199-207 (1997).

184. Bamberg, C.E., et al. The C5a receptor (C5aR) C5L2 is a modulator of C5aR-mediated

signal transduction. J Biol Chem 285, 7633-7644 (2010).

185. Settmacher, B., et al. Modulation of C3a activity: internalization of the human C3a

receptor and its inhibition by C5a. J Immunol 162, 7409-7416 (1999).

186. Naik, N., Giannini, E., Brouchon, L. & Boulay, F. Internalization and recycling of the

C5a anaphylatoxin receptor: evidence that the agonist-mediated internalization is

modulated by phosphorylation of the C-terminal domain. J Cell Sci 110 ( Pt 19), 2381-

2390 (1997).

187. Borregaard, N. & Cowland, J.B. Granules of the human neutrophilic polymorphonuclear

leukocyte. Blood 89, 3503-3521 (1997).

188. Borregaard, N., et al. Changes in subcellular localization and surface expression of L-

selectin, alkaline phosphatase, and Mac-1 in human neutrophils during stimulation with

inflammatory mediators. J Leukoc Biol 56, 80-87 (1994).

189. Sengelov, H. Complement receptors in neutrophils. Crit Rev Immunol 15, 107-131

(1995).

190. Gasque, P. Complement: a unique innate immune sensor for danger signals. Mol

Immunol 41, 1089-1098 (2004).

191. Guan, E., Robinson, S.L., Goodman, E.B. & Tenner, A.J. Cell-surface protein identified

on phagocytic cells modulates the C1q-mediated enhancement of phagocytosis. J

Immunol 152, 4005-4016 (1994).

192. Cotter, M.J., Zaiss, A.K. & Muruve, D.A. Neutrophils interact with adenovirus vectors

via Fc receptors and complement receptor 1. J Virol 79, 14622-14631 (2005).

193. Pliyev, B.K., Arefieva, T.I. & Menshikov, M.Y. Urokinase receptor (uPAR) regulates

complement receptor 3 (CR3)-mediated neutrophil phagocytosis. Biochem Biophys Res

Commun 397, 277-282 (2010).

Page 114: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

100

194. Losse, J., Zipfel, P.F. & Jozsi, M. Factor H and factor H-related protein 1 bind to human

neutrophils via complement receptor 3, mediate attachment to Candida albicans, and

enhance neutrophil antimicrobial activity. J Immunol 184, 912-921 (2010).

195. Amara, U., et al. Early expression changes of complement regulatory proteins and C5A

receptor (CD88) on leukocytes after multiple injury in humans. Shock 33, 568-575

(2010).

196. Shingu, M., et al. Activation of complement in normal serum by hydrogen peroxide and

hydrogen peroxide-related oxygen radicals produced by activated neutrophils. Clin Exp

Immunol 90, 72-78 (1992).

197. Vogt, W. Complement activation by myeloperoxidase products released from stimulated

human polymorphonuclear leukocytes. Immunobiology 195, 334-346 (1996).

198. Wirthmueller, U., et al. Properdin, a positive regulator of complement activation, is

released from secondary granules of stimulated peripheral blood neutrophils. J Immunol

158, 4444-4451 (1997).

199. Camous, L., et al. Complement alternative pathway acts as a positive feedback

amplification of neutrophil activation. Blood 117, 1340-1349 (2011).

200. Nauta, A.J., et al. Mannose-binding lectin engagement with late apoptotic and necrotic

cells. Eur J Immunol 33, 2853-2863 (2003).

201. Trouw, L.A., et al. C4b-binding protein and factor H compensate for the loss of

membrane-bound complement inhibitors to protect apoptotic cells against excessive

complement attack. J Biol Chem 282, 28540-28548 (2007).

202. Trouw, L.A., Nilsson, S.C., Goncalves, I., Landberg, G. & Blom, A.M. C4b-binding

protein binds to necrotic cells and DNA, limiting DNA release and inhibiting

complement activation. J Exp Med 201, 1937-1948 (2005).

203. Liszewski, M.K., Farries, T.C., Lublin, D.M., Rooney, I.A. & Atkinson, J.P. Control of

the complement system. Adv Immunol 61, 201-283 (1996).

204. Hogasen, A.K., Wurzner, R., Abrahamsen, T.G. & Dierich, M.P. Human

polymorphonuclear leukocytes store large amounts of terminal complement components

C7 and C6, which may be released on stimulation. J Immunol 154, 4734-4740 (1995).

205. Volanakis, J.E., Barnum, S.R. & Kilpatrick, J.M. Purification and properties of human

factor D. Methods Enzymol 223, 82-97 (1993).

206. Zipfel, P.F., et al. Factor H and disease: a complement regulator affects vital body

functions. Mol Immunol 36, 241-248 (1999).

207. Rifat, S., et al. Expression of sialyltransferase activity on intact human neutrophils. J

Leukoc Biol 84, 1075-1081 (2008).

Page 115: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

101

208. Sakarya, S., et al. Mobilization of neutrophil sialidase activity desialylates the pulmonary

vascular endothelial surface and increases resting neutrophil adhesion to and migration

across the endothelium. Glycobiology 14, 481-494 (2004).

209. Sengelov, H., Kjeldsen, L. & Borregaard, N. Control of exocytosis in early neutrophil

activation. J Immunol 150, 1535-1543 (1993).

210. Remijsen, Q., et al. Dying for a cause: NETosis, mechanisms behind an antimicrobial cell

death modality. Cell Death Differ 18, 581-588 (2011).

211. Pangburn, M.K., et al. Polyanion-induced self-association of complement factor H. J

Immunol 182, 1061-1068 (2009).

212. Heintzelman, D.L., Lotan, R. & Richards-Kortum, R.R. Characterization of the

autofluorescence of polymorphonuclear leukocytes, mononuclear leukocytes and cervical

epithelial cancer cells for improved spectroscopic discrimination of inflammation from

dysplasia. Photochem Photobiol 71, 327-332 (2000).

213. Liang, B. & Petty, H.R. Imaging neutrophil activation: analysis of the translocation and

utilization of NAD(P)H-associated autofluorescence during antibody-dependent target

oxidation. J Cell Physiol 152, 145-156 (1992).

214. Kim, Y.S., Park, H.H., Rhee, H.W., Hong, J.I. & Han, K. Neutrophils with toxic

granulation show high fluorescence with bis(Zn2+-dipicolylamine) complex. Ann Clin

Lab Sci 39, 114-119 (2009).

215. Heinen, S., Pluthero, F.G., van Eimeren, V.F., Quaggin, S.E. & Licht, C. Monitoring and

modeling treatment of atypical hemolytic uremic syndrome. Mol Immunol 54, 84-88

(2013).

216. Barnes, R.J., Leung, K.T., Schraft, H. & Ulanova, M. Chromosomal gfp labelling of

Pseudomonas aeruginosa using a mini-Tn7 transposon: application for studies of

bacteria-host interactions. Can J Microbiol 54, 48-57 (2008).

217. Botto, M., Lissandrini, D., Sorio, C. & Walport, M.J. Biosynthesis and secretion of

complement component (C3) by activated human polymorphonuclear leukocytes. J

Immunol 149, 1348-1355 (1992).

218. Hao, J., Meng, L.Q., Xu, P.C., Chen, M. & Zhao, M.H. p38MAPK, ERK and PI3K

signaling pathways are involved in C5a-primed neutrophils for ANCA-mediated

activation. PLoS One 7, e38317 (2012).

219. Guthrie, L.A., McPhail, L.C., Henson, P.M. & Johnston, R.B., Jr. Priming of neutrophils

for enhanced release of oxygen metabolites by bacterial lipopolysaccharide. Evidence for

increased activity of the superoxide-producing enzyme. J Exp Med 160, 1656-1671

(1984).

220. Schreiber, A., et al. C5a receptor mediates neutrophil activation and ANCA-induced

glomerulonephritis. J Am Soc Nephrol 20, 289-298 (2009).

Page 116: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

102

221. Kjeldsen, L., Bjerrum, O.W., Askaa, J. & Borregaard, N. Subcellular localization and

release of human neutrophil gelatinase, confirming the existence of separate gelatinase-

containing granules. Biochem J 287 ( Pt 2), 603-610 (1992).

222. Lipuma, J.J. The changing microbial epidemiology in cystic fibrosis. Clin Microbiol Rev

23, 299-323 (2010).

223. de Bentzmann, S. & Plesiat, P. The Pseudomonas aeruginosa opportunistic pathogen and

human infections. Environ Microbiol 13, 1655-1665 (2011).

224. van Hal, S.J., et al. Predictors of mortality in Staphylococcus aureus Bacteremia. Clin

Microbiol Rev 25, 362-386 (2012).

225. Martin-Ramirez, J., Hofman, M., van den Biggelaar, M., Hebbel, R.P. & Voorberg, J.

Establishment of outgrowth endothelial cells from peripheral blood. Nat Protoc 7, 1709-

1715 (2012).

226. Kennedy, S.A., Scaife, C., Dunn, M.J., Wood, A.E. & Watson, R.W. Benefits of heat

treatment to the protease packed neutrophil for proteome analysis: halting protein

degradation. Proteomics 11, 2560-2564 (2011).

Page 117: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

103

8 Appendix

Appendix A. Optimizing the Protocol for Neutrophil Lysates

When I first embarked on this project, one of the greatest challenges was to prepare a neutrophil

lysate. The trouble with preparing a neutrophil lysate arises from the neutrophil granules which

are very abundant in proteases. In addition, neutrophil myeloperoxidase is able to produce

hypochlorous acid, thus creating an environment that is unfavorable for recovering proteins.

In the literature, neutrophil research is often conducted with microscopy or flow

cytometry. For molecular work, neutrophil proteins are usually collected using a lysis buffer,

however the contents of the lysis buffer are often not disclosed. My first attempts to prepare a

neutrophil lysate using a lysis buffer containing 1% (v/v) Triton X-100 supplemented with 1 ×

cOmplete, mini protease inhibitor cocktail (Roche Diagnostics, Laval, QC, Canada) were

unsuccessful. In addition, heat treatment to prevent protein degradation was also unsuccessful226

.

While I was able to detect CFP in lysates, I was not able to detect loading controls such as β-

actin or GAPDH through western blot.

Moving forward, with advice from Dr. Lisa Robinson (The Hospital for Sick Children) I

was able to successfully prepare my first neutrophil lysate using a lysis buffer containing 1%

(v/v) Triton X-100, 45mM Tris-HCL pH 7.4, 120 mM NaCl, 1mM PMSF, 1mM Na3VO4, 2mM

PMSF and 2 × cOmplete, mini protease inhibitor cocktail. Neutrophils were lysed on ice for 10

minutes followed by sonication. To evaluate the efficiency of the different methods for preparing

neutrophil lysates, samples were separated with 10% (w/v) SDS-PAGE and proteins were

stained with Coomassie Brilliant Blue (Figure A1).

Page 118: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

104

Figure A1. Evaluating different methods for preparing neutrophil lysates.

Coomassie Blue staining was used to determine the most effective method for preparing

neutrophil lysates. Neutrophils were isolated from peripheral whole blood and lysates were

prepared using (A) 1% (v/v) Triton X-100 + 1 × cOmplete, mini protease inhibitor cocktail. (B)

1% (v/v) Triton X-100 + 1 × cOmplete, mini protease inhibitor cocktail heat treated at 95°C. (C)

1% (v/v) Triton X-100, 45mM Tris-HCL pH 7.4, 120 mM NaCl, 1mM PMSF, 1mM Na3VO4,

2mM PMSF and 2 × cOmplete, mini protease inhibitor cocktail. Samples were separated with

10% (w/v) SDS-PAGE.

Page 119: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

105

To further validate that this method for preparing neutrophil lysates was effective, I

utilized immunoblot analysis to detect common proteins that are used as loading controls.

Neutrophil lysates were separated with 10% (w/v) SDS-PAGE, transferred onto nitrocellulose

membrane. Western blot analysis was able to successfully detect myosin, β-actin, and GAPDH

in neutrophil lysates. In addition, we were able to identify the large membrane receptor CR1 in

neutrophil lysates (Figure A2).

Figure A2. Detection of proteins from neutrophil lysates. Neutrophil lysates were prepared and separated on 10% (w/v) SDS-PAGE. (A) Detection of

common loading controls, Myosin, β-actin, and GAPDH. (B) Detection of known neutrophil

membrane protein complement receptor 1.

The protocol for the preparation of neutrophil lysates has since evolved to what is now described

in this thesis (section 2.4) after continued consultation with literature and discussions with

experts in neutrophil biology at the international symposium on The Neutrophil in Immunity

(Quebec City, QC, Canada).

Page 120: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

106

Appendix B. Hemolysis Assay Data

A rabbit erythrocyte hemolysis assay was used to determine complement activity. A serial

dilution of the monoclonal antibody to CFP was performed in 20% (v/v) plasma in AP buffer to

determine the effective concentration of antibody required to inhibit complement AP on rabbit

erythrocytes. The effective concentration of antibody required to inhibit complement AP on

rabbit erythrocytes was dependent on donor plasma and varied from 2-8 μg/ml. The hemolysis

assay was performed on three separate dates and raw data for hemolysis determined through end

point reading at 405 nm can be found in (Table B1). The values are normalized to total

hemolysis of 2.5 × 107 rabbit erythocytes (50 μl) in distilled water (Table B2). The plasma from

June 19, 2013 displayed decreased complement activity after blockade with 2 μg/ml of

monoclonal antibody to CFP. Plasma from June 20, 2013 and July 4, 2013 showed decreased

complement activity after 4 μg/ml of monoclonal antibody to CFP (Figure B1). The data and

figures presented in this thesis corresponds neutrophils and plasma collected from the donor on

July 4, 2013 where 4 μg/ml of monoclonal antibody to CFP could effectively stop complement

AP activity (Figure B1C).

Table B1. Hemolysis absorption at 405 nm. Raw data as determined through endpoint reading at 405 nm. Controls A: AP Buffer, B: Rabbit

erythrocyte in AP buffer, C: Total hemolysis, D: 20% (v/v) plasma in AP buffer

Controls mouse monoclonal anti Factor P (μg/ml)

Date: A B C D 8 4 2 1 0.5 0.25 0.125

June 19, 2013 0.039 0.336 2.324 1.574 N/A 0.735 0.805 1.209 1.403 1.324 1.369

June 20, 2013 0.036 0.142 2.545 2.034 0.586 0.839 1.73 1.875 1.831 2.031 2.306

July 4, 2013 0.043 0.143 2.912 3.136 0.438 0.572 2.769 2.797 2.82 3.5 2.716

Table B2. Hemolysis absorption normalized to total hemolysis. Hemolysis data normalized to total hemolysis of rabbit erythrocytes resuspended in distilled

water. Controls A: AP Buffer, B: Rabbit erythrocyte in AP buffer, C: Total hemolysis, D: 20%

(v/v) plasma in AP buffer

Controls mouse monoclonal anti Factor P (μg/ml)

Date: A B C D 8 4 2 1 0.5 0.25 0.125

June 19, 2013 0.017 0.145 1 0.677 N/A 0.316 0.346 0.520 0.604 0.570 0.589

June 20, 2013 0.014 0.056 1 0.799 0.252 0.330 0.680 0.737 0.719 0.798 0.906

July 4, 2013 0.015 0.049 1 1.077 0.172 0.196 0.951 0.961 0.968 1.202 0.933

Page 121: Properdin Binds Pseudomonas aeruginosa and is Required for ...€¦ · ii Properdin Binds Pseudomonas aeruginosa and is Required for Neutrophil Extracellular Trap Mediated Activation

107

Figure B1. Complement activity assay determined by rabbit erythrocyte hemolysis.

Activity of complement AP was determined using rabbit erythrocyte hemolysis. A serial dilution

was performed to determine the concentration of mouse monoclonal antibody to CFP required to

inhibit complement AP. Three independent experiments from dates (A) June 19, 2013 (B) June

20, 2013 (C) July 4, 2013.