distinct oral neutrophil subsets define health and disease ... · siavash hassanpour master of...

75
Distinct Oral Neutrophil Subsets Define Health and Disease States by Siavash Hassanpour A thesis submitted in conformity with the requirements for the degree of Master of Science Faculty of Dentistry University of Toronto © Copyright by Siavash Hassanpour 2017

Upload: others

Post on 05-Oct-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

Distinct Oral Neutrophil Subsets Define Health and Disease States

by

Siavash Hassanpour

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

Faculty of Dentistry University of Toronto

© Copyright by Siavash Hassanpour 2017

Page 2: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

ii

Distinct Oral Neutrophil Subsets Define Health and Disease

States

Siavash Hassanpour

Master of Science

Faculty of Dentistry

University of Toronto

2017

Abstract

Neutrophils exit the vasculature and swarm to sites of inflammation and infection. Neutrophils

are also actively recruited to the healthy periodontium without eliciting clinical signs of

inflammation, suggesting a unique immune surveillance neutrophil function. We hypothesized

that oral neutrophils in the healthy periodontium exist in a “para-inflammatory” state that allows

neutrophils to symbiotically interact with the commensal oral microflora. Using the principles of

immunophenotyping, we identified four distinct neutrophil subsets: resting circulatory

neutrophils, two para-inflammatory and one pro-inflammatory tissue neutrophil phenotypes in

the absence and presence of chronic inflammation, respectively. This work is the first to

comprehensively characterize healthy oral neutrophils and identity the para-inflammatory

neutrophil phenotype, thereby demonstrating that not all neutrophils trafficking through

periodontium are fully activated. In addition to establishing possible diagnostic and treatment

monitoring biomarkers, the oral tissue neutrophil phenotype model builds on existing literature

suggesting that the healthy periodontium may be in a para-inflammatory state.

Page 3: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

iii

Acknowledgments

I would like to start by thanking by wonderful and beautiful wife Kelley Hassanpour who has

endured a seemingly never ending (thirteen-year) journey. None of my accomplishments would

have been possible without her boundless love and support. Kelley, I want to thank you for being

a constant source of happiness and my greatest motivation. We finally did it!

I would like to thank my mentor and supervisor Dr. Michael Glogauer. In 2008, with my

rejection from dental school looming large over my head, Michael gave me an opportunity to

work towards a Master of Science Degree. His faith in me allowed me to not only gain my first

Master of Science degree in 2010 but also propelled me into dental school and later into the

graduate periodontology program. Michael, I want to thank you for always believing in me and

for pushing me to go further than I ever thought possible. The lessons that I have learned from

you will stay with me for a life time and I am forever grateful for the opportunities you have

given me. I would also like to extend a warm thanks to my advisory committee members Drs.

Howard Tenebaum and James Scholey who’s guidance and wisdom were instrumental in the

completion of this project.

I would like to thank my dear friends from the University of Toronto’s Dentistry Class of IT4.

Timur Shigapov, Shlomi Tamam, Nicholas Tong, Alice Chen and Emily Tichenoff, I want to

thank you all for helping me survive the rigors of dental school. I also want to thank all the

Glogauer lab members who have been an integral part of my journey. I want to thank Jan Kuiper,

Noah Fine, Chunxiang Sun, Yongqiang Wang, Roland Leung, Guy Aboodi, Carol Forster,

Michael Lim, Corneliu Sima, Morvarid Oveisi and Alon Borensetin. Thank you all for your help

and support through the years. A very special thank you to Flavia Lakschevtiz who has literally

been there with me every step of the way for the last six years.

Finally, I want to thank my parents, Shohre Mahdi and Hosein Hassanpour and my sister

Maryam Amini, who’s countless sacrifices have given me a once in a life time opportunity.

Thank you everyone!

Page 4: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

iv

Table of Contents

Contents

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

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

List of Tables ................................................................................................................................. vi

List of Figures ............................................................................................................................... vii

Abbreviations ............................................................................................................................... viii

Chapter 1: Introduction ................................................................................................................... 1

Chapter 2: Literature Review .......................................................................................................... 2

2.1 The Inflammatory Spectrum and Pathogenesis of Periodontitis ......................................... 2

2.1.1 The Inflammatory Spectrum ................................................................................... 2

2.1.2 Inflammation and Periodontitis ............................................................................... 3

2.1.3 Pathogenesis of Chronic Periodontitis .................................................................... 3

2.2 Neutrophil Homeostasis and Lifecycle ............................................................................... 5

2.2.1 Neutrophil Differentiation ...................................................................................... 5

2.2.2 Circulatory Neutrophils .......................................................................................... 6

2.2.3 Neutrophil Extravasation ........................................................................................ 7

2.2.4 Neutrophil Clearance .............................................................................................. 8

2.3 Neutrophil Function ............................................................................................................ 9

2.3.1 Neutrophil Granules ................................................................................................ 9

2.3.2 Neutrophil Mediated Bacterial Killing ................................................................. 11

2.4 Neutrophil Plasticity and Heterogeneity ........................................................................... 12

2.4.1 Neutrophil Subpopulations ................................................................................... 13

2.4.2 Identification and Characterization and Cellular Subpopulations ........................ 14

Page 5: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

v

2.5 Oral Neutrophils as a Unique Population of Neutrophils ................................................. 16

Statement of the Problem .............................................................................................................. 20

Chapter 3 : Materials and Methods ............................................................................................... 21

3.1 Human Subjects ................................................................................................................ 21

3.2 Sample Collection and Processing .................................................................................... 21

3.3 Electron Microscopy ......................................................................................................... 22

3.4 Histopaque Density Centrifugation ................................................................................... 22

3.5 Multicolor Flow Cytometry .............................................................................................. 23

3.6 ROS Assay ........................................................................................................................ 23

3.7 NET Assay ........................................................................................................................ 24

3.8 Statistical Analysis ............................................................................................................ 24

Chapter 4 : Results ........................................................................................................................ 25

4.1 Clinical Description of Periodontal Status ........................................................................ 25

4.2 CP Oral Neutrophils Are More Degranulated and Have Increased Phagocytosis

Compared to Healthy Oral Neutrophils ............................................................................ 25

4.3 Flow Cytometric Gating Strategy ..................................................................................... 26

4.4 Characterization of Oral Neutrophil Populations in Health and CP ................................. 26

4.5 Pro-inflammatory Neutrophils Have Elevated ROS Production and NET Formation ..... 27

Chapter 5 : Discussion .................................................................................................................. 28

Conclusion and Future Directions ................................................................................................ 31

Figure Legend ............................................................................................................................... 33

Tables and Figures ........................................................................................................................ 36

Contributions to the Thesis and Manuscript ................................................................................. 49

References ..................................................................................................................................... 50

Page 6: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

vi

List of Tables

Table 1: CD Marker Panels for Multicolor Flow Cytometry.

Table 2: Demographic and Periodontal Characteristics of Patients Cohorts.

Table 3: CD Marker Expression Levels on Healthy and CP Neutrophils.

Page 7: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

vii

List of Figures

Figure 1. Neutrophil Recruitment to the Periodontium in Health and Disease.

Figure 2. Etiology and Pathogenesis of Chronic Periodontitis.

Figure 3. Multicolor Flow Cytometry Gating Strategy for Blood and Oral Neutrophils.

Figure 4. Oral Neutrophils Have Elevated Phagocytosis and Greater Degranulation in CP.

Figure 5. Identification of Para-inflammatory Neutrophils in Oral Health.

Figure 6. Para- & Pro-inflammatory Oral Neutrophil Populations Are Defined by Unique CD

Marker Expression Profiles.

Figure 7. CD Markers Can Be Used to Gate on Para1 and Para2 Oral Neutrophil Populations.

Figure 8. Pro-inflammatory Oral Neutrophil Populations Have Elevated NET Formation and

Increased ROS Production Compared to Para-inflammatory Neutrophils.

Figure 9. Model of Para- and Pro-inflammatory Neutrophil Phenotypes in Health and Disease.

Page 8: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

viii

Abbreviations

AO – Antioxidant

BOP – Bleeding on Probing

CAL – Clinical Attachment Loss

CCL – Chemokine Ligand

CD – Cluster of Differentiation

CGD - Chronic Granulomatous Disease

CP – Chronic Periodontitis

CXCR - CXC Chemokine Receptor

DEL - Developmental Endothelial Locus

DHR - Dihydrorhodamine

DNA - Deoxyribose Nucleic Acid

E/A - Epon Araldite

FACS - Fluorescence-Activated Cell Sorting

fMLP – Formyl-methionyl-leucyl-phenylalanine

FMO – Fluorescence Minus One

FSC – Forward Scatter

GCF - Gingival Crevicular Fluid

G-CSF – Granulocyte - Colony Stimulating Factor

GI – Gastrointestinal

HD – High Density

HDN – High Density Neutrophils

HLA - Human Leukocyte Antigen

HTS - High-Throughput Screening

ICAM – Intercellular Adhesion Molecule

INF – Interferon

Page 9: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

ix

IL – Interleukin

LD – Low Density

LDN – Low Density Neutrophils

LFA- Leukocyte Function Associate Antigen

LXR – Liver X Receptor

NAPDH - Nicotinamide Adenine Dinucleotide Phosphate

NET – Neutrophil Extracellular Trap

NRF2 - Nuclear Factor Erythroid 2-Related Factor

MFI - Mean Fluorescence Intensity

MMP - Matrix Metalloproteinases

MPO - Myeloperoxidase

MRSA - Methicillin-Resistant Staphylococcus aureus

PARA – Para-inflammatory

PBS – Phosphate-Buffered Saline

PD – Probing Depth

pDC - Plasmacytoid Dendritic Cells

PFA – Paraformaldehyde

PKC – Protein Kinase C

PMA - Phorbol 12-Myristate 13-Acetate

PMN – Polymorphonuclear Cells or Neutrophils

PO - Propylene Oxide

PRO – Pro-inflammatory

RNA – Ribonucleic Acid

ROS - Reactive Oxygen Species

SSC – Side Scatter

SLE - Systemic Lupus Erythematous

Page 10: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

x

TAN - Tumor Associated Neutrophils

TNF - Tumor Necrosis Factor

TLR - Toll-Like Receptors

VAMP - Vesicle Associated Membrane Protein

Page 11: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

1

Chapter 1: Introduction

Neutrophils are essential primary innate immune responders that kill invading pathogens, but can

also cause substantial host tissue damage as part of the inflammatory response to those pathogens

[1]. Chronic periodontitis (CP), a highly prevalent, dysbiotic, inflammatory condition of the oral

cavity, is a model of tissue destruction mediated by the pro-inflammatory neutrophil response [2-

7]. CP is characterized by amplified neutrophil recruitment [8], gingival inflammation and

progressive, irreversible loss of periodontal attachment. Paradoxically, neutrophils are also

constitutively recruited to the oral cavity in health without inducing clinically evident

inflammation or tissue destruction [9,10] (Figure 1). Within tissues, the intermediary immune

state that allows the host to adequately respond to low grade noxious agents or tissue damage,

without clinical signs of inflammation, has been termed “Para-inflammation” [11-14].

Immunogenic events that induce a switch from a para-inflammatory to a pro-inflammatory state

are likely to be central to the pathogenesis of chronic inflammatory diseases associated with

biofilm-bearing tissues. Mechanisms that restrain oral neutrophil function in health and allow for

management and tolerance of the commensal microbiota while avoiding tissue damage may be

essential regulators of the para-inflammatory state. Normal circulating blood neutrophils are

naïve, becoming primed or activated during certain disease states, while oral neutrophils, which

have undergone extravasation and exposure to the oral biofilm, are activated [15]. Based on

reports of characteristic oral neutrophil phenotypes associated with periodontitis [16] and

neutrophil phenotype heterogeneity in other pathological conditions [17-19], we hypothesized

that oral neutrophils associated with periodontal health might have a distinct phenotype from

neutrophils associated with inflamed periodontal tissues. Using CP, a model of oral mucosal and

connective tissue inflammation, in conjunction with comprehensive cluster of differentiation

(CD) marker expression profile analysis we demonstrate that two unique tissue neutrophil

phenotypes are present in the oral cavity: a Para-inflammatory neutrophil in the healthy oral

cavity, and a Pro-inflammatory neutrophil, which occurs in CP.

Page 12: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

2

Chapter 2: Literature Review

2.1 The Inflammatory Spectrum and Pathogenesis of Periodontitis

2.1.1 The Inflammatory Spectrum

Inflammation is the adaptive response to a noxious stimulus. At its core, the inflammatory

machinery can be subdivided into four basic components: a stimulus, a sensor, a mediator and

the target tissue(s). Bacterial, fungal, parasitic or viral infections as well as non-infected tissue

injury all trigger unique sets of events aimed at controlling and eliminating the source of noxious

stimulus. Acute inflammation is resolved with the elimination of the triggering stimulus, repair

of damaged target and reestablishment of homeostasis [20]. The tapering and cessation of acute

inflammation is a regulated mechanism that involves a transition from pro-inflammatory to pro-

resolution mediators as well as a shift from neutrophil to macrophages as the predominant

inflammatory cell[21]. If acute inflammation is unresolved either through the persistence of the

triggering stimulus or the inability to resolve the acute inflammatory response, chronic

inflammation ensues resulting in tissue damage. The transition from the protective role of acute

inflammation into the damaging effects of chronic inflammation is the reason why inflammation

is associated with a myriad systemic disorders such as obesity, diabetes, cancer and

cardiovascular disease [20]. Para-inflammation, is an intermediary inflammatory state that allows

host tissues to adapt and tolerate a mild noxious stimulus with the physiological role of

maintaining or restoring tissue function and homeostasis [22]. The evolutionary origins of Para-

inflammation are though to arise from exposure to stimuli that were not present during early

human history such as over-feeding and aging [14]. The key mediator of para-inflammation in

the peripheral tissues are resident macrophages but depending on the severity of the stressor

stimulus can include other resident cells (e.g. mast cells and endothelial cells) and recruit

circulatory inflammatory cells (e.g. inflammatory monocytes). Much like unresolved acute

inflammation, unresolved chronic para-inflammation results in initiation and progression

pathological conditions such as type 2 diabetes mellitus, atherosclerosis and age-related

neurodegenerative diseases [14,23]. There is currently no comprehensive explanation of the

mechanisms that lead to an excessive, dysregulated inflammatory response that from shifts the

Page 13: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

3

beneficial reparative functions of inflammation into detrimental tissue damage role of

inflammation.

2.1.2 Inflammation and Periodontitis

The protective yet destruction duality of inflammation is best illustrated by the neutrophil

paradox in periodontal health and disease. The oral cavity is colonized early in life with plethora

of microbes [24]. Early colonization of the oral cavity allows for the induction and maintenance

of immunological microbial tolerance for the host. Microbial tolerance is crucial for our survival

considering each human cell in our body is out-numbered by ten microbial cells [25]. Our ability

to amicably tolerate a vastly diverse commensal flora suggests that our immune system is

capable of sensing beneficial microorganisms, while targeting and eliminating pathogenic

microbial organisms by activating the innate and adaptive immune system [26]. In return,

commensal organisms provide the host with unevolved traits and instruct and guide the proper

induction and functioning of the immunological system [27]. The major players in the sensing

and protection of commensal organisms have been reported to be gut epithelial cells, dendritic

cells and macrophages [28][29] [27]. The oral cavity, a direct extension of the GI is dominated

by a single myeloid lineage, the oral neutrophil. Despite the lack of the macrophages and

dendritic cells, the oral neutrophils are still capable of coexisting with the healthy commensal

oral microbiota without eliciting a true an inflammatory reaction, suggesting a previously

unreported immune surveillance role for the oral neutrophil.

2.1.3 Pathogenesis of Chronic Periodontitis

Periodontitis is a chronic inflammatory disorder that leads to the destruction of the supporting

apparatus of teeth (the periodontium), resulting in progressive loss of dental attachment that

culminates in the premature exfoliation of teeth. Periodontitis is global problem that affects

nearly half of the world’s population. In North America, 46% of the population has periodontitis,

with 8.9% of the populations suffering from severe chronic periodontitis [3]. The etiology of

periodontitis is multifactorial and relies on the microbial, host and systemic factors (Figure 2).

At the microbial level, periodontitis pathogenesis depends on the quality and quantity of the

microbial biofilm. Classically, the “red complex” bacteria (Porphyromonas gingivalis,

Page 14: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

4

Treponema denticola and Tannerella forsythia) were viewed as the causative agent of

periodontitis [30]. However, recent evidence suggests that the microbial pathogenesis of

periodontal disease goes beyond the mere presence of the “red complex” microbial species. In

fact, periodontal infections are rarely caused by a large biomass of a solitary specie. Rather,

complex, multi-species microbial populations called biofilms induce most oral and periodontal

infections. Therefore, the etiology of periodontitis on a microbial level periodontitis is best

described by an micro-environmental shift that disrupts a healthy and harmonious (“symbiotic”)

biofilm into a pathogenic and destructive (“dysbiotic”) microbial community [31-33]. In

periodontal health, there exists an equilibrium between commensal microorganisms and the host

immuno-inflammatory state. This is analogous to the harmonious relationship between the

commensal gut microbiota and mucosal immune cells that allows for a state of “controlled

inflammation” in the intestines [34]. By quickly colonizing multiple oral niches such as the

tongue, teeth and gingival sulcus, the symbiotic microbiome confers colonization resistance to

the host by impeding the colonization of oral niches with pathogenic bacteria. Further, the

symbiotic periodontal microbiota, composed primarily of facultative gram positive bacteria of

the Actinomyces and Streptococci species, protects the host by promoting proper tissue structure

and function [35], partly by regulating and shaping the mucosal immune response[35,36]. This is

in stark contrast to the dysbiotic microbial community which is predominated by gram negative,

anaerobic microorganisms rich with virulence factors that allow the biofilm circumvent the

adaptive and innate immune system thus allowing the pathogenic microbiota to flourish in a pro-

inflammatory environment[31-33]. In a dysbiotic biofilm, pathogenic bacteria, such as P.

gingivalis, subvert the immune system and induce major qualitative and quantitative changes in

the oral biofilm, triggering a robust inflammatory response that ultimately results in host tissue

damage [37].

Bacterial dysbiosis may be required to initiate periodontitis but dysbiosis alone often fails to

cause prolonged attachment loss. Many individuals who lack adequate professional and personal

sanative care present with heavy supra and sub-gingival dysbiotic biofilms fail to display the

clinical signs and symptoms of periodontal destruction. These individuals appear to tolerate the

dysbiosis due to a hypo-responsive immune-inflammatory reaction. Conversely, other members

of the population present with severe inflammatory response in the face of a slight pathological

Page 15: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

5

microbial shifts[38]. Although certain behavioral (e.g. smoking) [39], systemic (e.g. uncontrolled

diabetes) [40] and genetic (e.g Interleukin (IL)-1 polymorphism) [41] risk factors have been

described , nearly half of the variation in periodontal disease susceptibility is unaccounted for

[42]. It is postulated that some individuals may be less susceptible to periodontal disease and

have an inherent ability to resist the conversion of a symbiotic microbiota into a dysbiotic one

thanks to a yet to be identified undetermined quality of the immune system. We have proposed

that the missing susceptibility link in periodontitis susceptibility lies within the oral neutrophil

phenotypes of patients [43,44].

2.2 Neutrophil Homeostasis and Lifecycle

2.2.1 Neutrophil Differentiation

To better appreciate the role of neutrophils in the pathogenesis and resistance to periodontitis, it

is beneficial to first understand the neutrophil homoeostasis and function. Neutrophil

homoeostasis is a well-tuned machine with multiple mechanisms that tightly regulate neutrophil

production, trafficking and clearance [45]. Neutrophils are terminally differentiated, non-

proliferating cells that account for two-thirds of all leukocytes in blood [46]. This impressive

number of circulatory neutrophils is sustained through the daily differentiation of over 1011

neutrophils in the bone marrow [47][46]. Neutrophil differentiation can be divided into six stages

based on the basis of cell size, nuclear morphology, granule content and mitotic activity

(Reviewed in detail [48]). Neutrophil production and egress from the bone marrow is primary

controlled through the granulocyte colony-stimulating factor (G-CSF), as demonstrated by severe

neutropenia in animal models and human patients lacking the G-CSF receptor [49]. G-CSF hones

neutrophil progenitors to the bone marrow, helps commit progenitors to the myeloid lineage and

controls the release of mature neutrophils to the blood stream [49]. Interleukin (IL) -17 can also

promote granulopoiesis and promote the recruitment, activation and survival of neutrophils by

upregulating G-CSF expression [46,50,51]. Neutrophil egress is also mediated through the

expression of CXC Chemokine Receptor (CXCR) 4 and CXCR2 receptors on neutrophil

precursors. Neutrophil maturation in the bone marrow coincides with a progressive and gradual

decline in CXCR4 expression that is counterbalanced by an increased expression of CXCR2

expression in the latter stages of neutrophil maturation [52]. CXCR4 receptor signaling is

responsible for the initial recruitment of neutrophil progenitors to the bone marrow, while

Page 16: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

6

continued expression of CXCR4 receptor is required for the retention of the neutrophil

precursors in the bone marrow. The deletion of the CXCR4 gene results in a marked egress of

neutrophils into circulation from the bone marrow [53][54][1][55,56]. Immune cells can also

directly influence the egress of neutrophils from the bone marrow. For instance, dendritic cells

can influence the distribution of neutrophils between the bone marrow, peripheral organs and

blood [1], while phagocytosis of apoptotic neutrophils by macrophages results in a negative

feedback loop that depresses the production of the pro-inflammatory cytokine IL-23, which in

turn decreases the production of IL-17 by T-lymphocytes. A drop in IL-17 ultimately decreases

the production of G-CSF by fibroblasts, thus limiting the egress of neutrophil from the bone

marrow [57,58].

2.2.2 Circulatory Neutrophils

Once fully formed, mature neutrophils are released into the blood stream and recruited to tissues

to ward off infections and resolve tissue damage. Mature circulatory neutrophils possess the

entire complement of antimicrobial factors, respiratory burst machinery and receptors required to

mount an inflammatory response prior their egress from the bone marrow (see below). Mature

neutrophils have a limited lifespan in circulation that typically requires little to no functional

activity. To that end, circulatory neutrophils are largely considered to be “dormant” cells that

don’t readily respond to activating stimuli [46]. This dormancy should not be equated to

transcriptional inactivity as studies have shown that circulatory neutrophils constitutively express

over 12,000 transcripts, while other studies have shown mature circulatory neutrophils to

upregulate the expression of several transcription factors and increase ribonucleic acid (RNA)

biosynthesis in response to inflammatory challenges. [46,59].

Under physiological conditions, only mature neutrophils are released into circulation such that

immature neutrophil phenotypes represents less than three percent circulatory neutrophils [60].

Neutrophilia, induced by acute inflammation accelerates neutrophil recruitment from the bone

marrow, resulting in the depletion neutrophil reserves in the bone marrow and marginated pools

in the lungs, live and spleen [1][61,62]. To compensate for the increase in demand, neutrophil

maturation time is reduced resulting in the release of functionally competent yet morphologically

immature neutrophils [63]. This is supported by the presence of CD177+, immature neutrophils at

Page 17: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

7

sites of infection, presumably to assist in the local production of neutrophils and help in the

clearance of severe infections [64]. This process is most likely regulated through “emergency

granulopoiesis”. Unlike steady state granulopoiesis, emergency granulopoiesis is initiated by

microbial challenges, bacterial products such as endotoxins, inflammatory mediators such as IL-

1, Tumor Necrosis Factor (TNF)- and G-CSF [65]. Neutrophils with immature phenotype have

also been described in a number of pathological and physiological conditions ranging from SLE,

cancer, sepsis [17,18].

2.2.3 Neutrophil Extravasation

When a microbial challenge breaches the local physical barriers, neutrophils aggregate around

the infection in multiple waves. Rapid recruitment of neutrophils to tissues requires the

coordinated mobilization of neutrophils from bone marrow reserves, accelerated hematopoiesis

and recruitment from marginated pools [66,67]. Neutrophil extravasation is a cascading, stepwise

process that involves rolling, firm adhesion, polarization, and transmigration[47]. Neutrophil

rolling is the initial interaction between granulocytes and vascular endothelial cells and is

mediated through a family of C-type lectin glycoproteins known as selectins [68]. L-selectin

(CD62L) is key in regulating neutrophil rolling and is constitutively expressed on the neutrophil

membranes. The interaction of CD62L with its ligand on the surface of endothelial cells initiates

a low-affinity interaction that tethers and slows circulating neutrophils into “rolling” along the

vascular endothelial cells [69]. Selectin mediated bonds are weak and transient in nature but are

sufficient to activate and polarize captured neutrophils to activate a second phase of higher

affinity interactions mediated by Leukocyte Function Associate Antigen (LFA)-1 (CD11a/CD18)

neutrophil integrins and endothelial cell Intracellular Adhesion Molecule-1 (ICAM-1) [70,71].

This interaction can be inhibited by a recently identified glycoprotein, Developmental

Endothelial Locus (DEL)-1 thus suppressing firm adhesion and subsequent transmigration [72].

While in circulation, neutrophils only express relatively few low-affinity β2-integrins. However,

upon contact with the endothelial membrane 2-integrins quickly transform into a high affinity

or active state and increase dramatically in number and membrane density. Once activated

integrins mediate adhesion and outside-in signaling that culminate in changes in the actin

cytoskeleton at the leading edge of the cell that mediate neutrophil transmigration [73]. Once

firmly attached to the endothelial lining, neutrophils begin the process of transmigration out of

Page 18: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

8

the vessel lumen and into the underlying tissue either between endothelial cells or through them

[74]. During transmigration, neutrophils dynamically alter their membrane phenotype by

initiating the exocytosis of secretory vesicles that contain CD11, CD18, CD31, CD54, CD44 and

CD47 to the cell surface [1,46,75-79]. The transmigration cascade culminates as the elongated

uropod of the emigrating neutrophil detaches from the vessel wall and the neutrophil migrates

into tissue [80].

As neutrophils leave the circulation they must adapt and transform to suit their new function as

tissue phagocytes. To this end, emigration from circulation alters the functional state of

neutrophils characterized by enhanced Nicotinamide Adenine Dinucleotide Phosphate (NAPDH)

oxidase activity [81], increased production of pro-inflammatory cytokines such as IL-8 [82] and

enhanced anti-apoptosis mechanisms. In addition, tissue neutrophils are also more

transcriptionally active than circulatory neutrophils[16]. The burst of transcriptional activity in

tissue neutrophils promotes the antimicrobial capacity of neutrophils and generates and promotes

the release of immune active chemokines and cytokines that recruit additional inflammatory cells

[82]. The transition from a passive, dormant circulatory neutrophil to a more transcriptionally

active, antimicrobial, immune active tissue neutrophil is commonly referred to as activation.

2.2.4 Neutrophil Clearance

As neutrophils continue to age in circulation without being targeted to tissues a feedback circuit

involving L-selectin, chemokine receptors and integrins are activated [1]. Aging circulatory

neutrophils shed L-selectin (CD62L), upregulate the expression of the bone marrow homing

receptor CXCR4 as well as CD11b. Collectively these changes target old circulatory neutrophils

(CD62Llo, CXCR4hi) back to the bone marrow where they are cleared by macrophages. This

processes promotes the activation of Liver X Receptor (LXR) transcription factors in

macrophages, which in turn increases the expression of CXCL12 in the bone marrow. Enhanced

CXCL12 expression promotes the release of newly formed mature neutrophils (CD62Lhi,

CXCR4lo) back into circulation. This process repeats in a oscillating circadian rhythm and

ensures an equilibrium state between the introduction of newly formed neutrophils and removal

of aged neutrophils into and out of circulation [83]. Aging is accompanied by depressed

neutrophil function. Studies in elderly humans have shown that neutrophils maintain their

Page 19: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

9

chemotactic ability but are inefficient in bacterial clearance and cause more host mediated tissue

destruction through the inadvertent spilling of protease contents [1].

Controlled neutrophil clearance is essential for the maintenance of tissue hemostasis and

resolution of inflammation. Deficiencies in controlled neutrophil clearance results inappropriate

neutrophil activation, in the progression of chronic inflammation [84,85] and ultimately ends in

host tissue destruction. Neutrophil clearance occurs either through programed cell death

(apoptosis) or retreat from inflammatory sites against the chemotactic gradient (reverse

migration) [21][86]. Once at the site of inflammation, neutrophils normally undergo apoptosis

after engaging the offending insult. However, recent evidence suggests neutrophils are capable of

migration against the chemoattractant gradient. Reverse neutrophil transmigration has been

shown to be critically important in inflammation resolution. The exact pathophysiology and

implications have yet to be fully elucidate [86]. Apoptosis on the other hand, allows for the

clearance of neutrophils while minimizing the release of cytotoxic and proteolytic granule

contents to the surrounding tissues[87]. Apoptosis is regulated through the engagement of death

receptors found on the surface of apoptotic neutrophils. During the early stages of neutrophil

maturation, apoptosis is only triggered in response to aberrant Deoxyribose Nucleic Acid (DNA)

replication. However, as neutrophils mature they pack their secretory vesicles with so called

death receptors that respond to extracellular death ligands commonly found in pro inflammatory

microenvironment [1]. To prolong their survival outside the bone marrow, neutrophils become

anti-apoptotic in response to various growth factors and cytokines and thus earn prolonged life

span following the egress from the bone marrow and during diapedesis [88].

2.3 Neutrophil Function

2.3.1 Neutrophil Granules

Throughout neutrophil maturation, undifferentiated cells acquire morphological and functional

features necessary for the neutrophil antimicrobial activity. During neutrophil development,

granules first appear in the transition from myeloblast to promyelocyte stage. Structurally, four

granule subsets, including azurophil (primary), specific (secondary), gelatinase (tertiary) and

secretory vesicles have been described in the literature [89]. Azurophil granule protein contents

Page 20: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

10

are formed early during the promyelocyte stage. Azurophil granules are the lysosomes of

neutrophils and contain myeloperoxidase (MPO) as well as defensins and acid hydrolases.

Specific granules contents are synthesized at the myelocyte stage and consists of bacteriostatic

and bactericidal agents including proteolytic enzymes and complement activators. As their name

suggests, gelatinase granules are packed with gelatinases, phosphates and Matrix

Metalloproteinases (MMPs). Gelatinase granule contents are formed at the metamylelocyte stage

and neutrophils use their targeted release to facilitate tissue migration. Secretory vesicles are the

last neutrophil granules to be formed and are the main source of membrane receptors and CD

markers such as CD11, CD18, CD67, CD14, CD35, CD10 and CD13 [89,90]. Unlike other

granule subsets, the key to secretory vesicles lies within the membranes as opposed to the

granule contents. Again, unlike other granules, the release of secretary vesicles is not only

regulated by inflammatory signals such as IL-8 and Formylmethionyl-leucyl-phenylalanine

(fMLP) [55,90,91] but by transmigration via interaction of L-selectin on circulatory neutrophils

and endothelial P- selectin [55,92]. As secretory vesicles fuse with the plasma membrane, they

“prime” the neutrophil for antimicrobial activity by delivering a rich source of receptors and

other functional proteins to the neutrophil surface. Importantly, the fusion of secretory vesicles

enhances neutrophil functional capacity without releasing proteolytic and cytolytic granule

content[89]. Overall, neutrophil granules display a continuum with respect to their propensity for

exocytosis that is best described by the targeting-by-timing hypothesis [93]. The enhanced

exocytosis capabilities of gelatinase and secretory vesicles compared to specific and azurophil

granules is directly correlated to density of expression of the fusogenic proteins such as Vesicle

Associated Membrane Protein (VAMP)-2 on the granule membrane surface [94]. Following a

mild trigger, such as a transient rise in intracellular calcium ions, it is detrimental to neutrophils

and the host to indiscriminately and simultaneously release all granule contents. It is more

prudent for neutrophils to grade their response according to the situation. When neutrophils

initially transmigrate out of circulation and into the tissues, there is a specific need for the

targeted release of gelatinizes, MMPs, and proteases. This initial of matrix degradation

supersedes the need for complement receptors that mediate phagocytosis of microorganisms or

pro-antibacterial proteins. To that end secretory and gelatinase granules with the highest

concentration of VAMP-2 are released first. Only a sustained exocytic stimulation allows for the

release of the tissue destructive specific and azurophil granules [89].

Page 21: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

11

2.3.2 Neutrophil Mediated Bacterial Killing

Neutrophils neutralize invading microorganisms through a three-pronged approach consisting of

an oxygen dependent pathway, an oxygen independent pathway and NETosis. The majority of

the antimicrobial activity of neutrophils occurs within phagosomes. During phagocytosis,

opsonized targets are ingested through the generation and extension of pseudopods that

encapsulate the organism [95]. The internalized particles are housed in membrane-bound

phagosomes, which fuse with granules containing proteolytic proteins [96]. Once ingested,

microbes trapped in phagosomes can be neutralized through the oxygen dependent pathway

revolving around the assembly of the multiprotein enzyme complex known as the reduced

NADPH oxidase. When activated neutrophils dramatically increase oxygen consumption and

activate the NADPH oxidase complex that is normally quiescent. Once the NADPH oxidase

complex is assembled, it functions as an electron transport chain that funnels negative charges

into the phagosome lumen and thereby depolarizes the phagosomal membrane, initiates the

respiratory burst and catalyzes the formation of superoxide anion [97][98]. The NAPDH oxidase

enzyme complex is composed of the oxidase proteins p22phox, p47hox, p67phox and gp91phox,

p40phox (suffix phox representing phagocyte oxidase) the GTPases Rac1/2 and Rap1A.

Activation of the electron transport machinery requires the translocation of cytoplasmic p47phox,

p67phox and p40phox to the phagosome membrane [99]. The importance of the NAPDH oxidase in

neutrophil function is best characterized by mutation in genes the encode the subunits of the

NAPDH oxidase collectively known as Chronic Granulomatous Disease (CGD) [100].

Neutrophils from CGD patients fail to produce normal amounts of superoxide. Superoxide (O2-)

is the primary product of the respiratory burst. Superoxide subsequently dismutates into

hydrogen peroxide (H2O2) [101]. Neutrophils can also release MPO which combines with H2O2

to produce the extremely cytotoxic hypoclorous acid. Superoxide and hydrogen peroxide impart

their cytotoxic effects thanks to their ability to generate additional Reactive Oxygen Species

(ROS) namely hydroxyl radicals (OH-), singlet oxygen (1O2) and ozone (O3). The generated

ROS contribute tissue destruction and pathogenesis of periodontal disease [102] and respiratory

levels are commonly used as an indicator of neutrophil activation [6].

Page 22: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

12

Neutrophil inflammatory response was largely believed to be intracellular. However, the

discovery of Neutrophil Extracellular Traps (NET), shed light on the extracellular contribution of

neutrophils in innate immunity. NETs are composed of strands of DNA wrapped in proteolytic

proteins that are capable of entrapping and killing invading microbes, thus minimizing the extent

or rate of spread of an infection [1][103]. The formation of NETs (or NETosis) is a terminal

event for neutrophils and is an alternative death pathway to apoptosis. NETosis begins as

neutrophils dissolve their nuclei, chromatin and granules. Neutrophils then extrude large strands

of histone covered DNA coated with cytosolic and granular proteins [104,105]. NET associated

proteins are categorized as either bactericidal (such as histones, defensins, lactoferrin, and MPO),

pattern recognition proteins (such as Pentraxin 3) or cytosolic proteins (calportectin) [47]. In an

in vitro setting, NETosis induced by Phorbol 12-Myristate 13-Acetate (PMA), is dependent on

the presence of elastase, MPO and active NAPDH oxidase [106]. As expected, NETosis is

devoid or absent in patients that suffer from conditions that cause a deficiency in MPO, an

inactive NADPH oxidase or the lack elastase and the serine proteases [107-109]. NETs may also

be a major contributory factor to the onset and exacerbation of a number of autoimmune

diseases. NETs contribute to autoimmune disease such as systemic lupus erythematous and

Wegener’s granulomatosis as they lead to the extracellular release of multiple intracellular target

antigens such as DNA and MPO [110].

2.4 Neutrophil Plasticity and Heterogeneity

Despite the reported complexity, neutrophils are often discounted as a phenotypically

homogenous population with a suicidal mentality of swarming sites of infection to exuberantly

clear out invading pathogens only to perish in the infiltrated tissues. However, there has been a

recent paradigm shift in the understanding and appreciation of neutrophil diversity. A growing

body of evidence suggests that neutrophils should be viewed as diverse population with a broad

spectrum of phenotypes and functions and that are subject to microenvironmental modulation.

Recent investigation into neutrophil biology as changing our understanding of every aspect

neutrophil. For instance, we now know that circulatory neutrophils are longer lived than

originally thought, with a reported circulatory half-life of over five days [111,112]. We also

know that aging neutrophils not recruited to sites infection display a unique phenotype

characterized by diminished functional capacity (degranulation, respiratory burst, phagocytosis,

Page 23: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

13

chemotaxis), increased expression of bone marrow homing signals, decreased sensitivity to

inflammatory signals and cytokines and increased apoptotic tendency [113]. Research has also

shown that neutrophils are not necessarily “dead end” cells and are capable of reverse

transmigration (migration from tissues – to blood). The process of reverse transmigration results

in distinct phenotypic changes in neutrophils characterized by increased expression of ICAM-1

and low expression of CXCR1. These ICAM-1highCXCR1low displayed reduced tendency to

migrate through an endothelial barrier, increased cell rigidity, increased ROS production and

decreased apoptosis. ICAM-1highCXCR1low neutrophils have been detected in the circulation of

patients with rheumatoid arthritis and atherosclerosis but their role in the pathogenesis of these

chronic inflammatory conditions has yet to be elucidated [86]. However, the process of reverse

transmigration does likely lead to the generation of potentially pathogenic subset of neutrophils

that may disseminate systematic inflammation from a localized site of injury to distant organs

[114]. Lastly, diverse and novel functions for neutrophils have been described in details in

complex interactions with the adaptive immune response (reviewed in detail in [115-117].

2.4.1 Neutrophil Subpopulations

Distinct neutrophil subpopulations have been identified in a number of pathological conditions.

In a murine cancer model, cancer was associated with specialized neutrophil subsets with a

unique transcriptional profile referred to as Tumor Associated Neutrophils (TAN) [118]. Within

the TAN family of neutrophils, two subsets were identified, namely N1 and N2 neutrophils or

PMNs. N1 neutrophils were created by the blockage of the transforming growth factor- (TGB-

) and had an immunostimulatory and anti-tumor phenotype. N2 neutrophils, which constituted

the majority of TANs, were in turn immunosuppressive and pro-tumorigenic. Two unique

neutrophil populations (PMN-I PMN-II) have also been identified in a murine Methicillin-

Resistant Staphylococcus aureus (MRSA) model [119]. PMN-I neutrophils are induced in

MRSA infected mice as a result of a mild secondary systemic inflammatory response while

PMN-II are induced in Methicillin-Resistant Staphylococcus aureus (MRSA) infected mice as a

result of a severe secondary inflammatory response. When compared to PMN-I neutrophils,

PMN-II neutrophils have a more immature nuclear morphology and express differing cytokines,

chemokines, Toll-Like Receptors (TLR) and adhesions molecules. These differences ultimately

lead to impaired activation of macrophages resulting in inefficient anti-bacterial response and

Page 24: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

14

clearance of MRSA [119]. Neutrophil heterogeneity has also been identified in sepsis. Not

surprisingly sepsis is associated with profound neutrophilia. However, despite the increase in

circulatory neutrophils, septic patients are severely immune compromised, prone to infections

and often succumb to their illness [120]. Decreased immunity in the face of neutrophilia is

suggestive of an altered function of circulating neutrophils. In response to systemic endotoxins, a

heterogeneous neutrophil population can be identified with an increase in the proportion of

neutrophils characterized by high levels of CD markers, CD16, CD11b and CD11c and low

levels of CD62L [121]. These cells displayed reduced interactions with opsonized bacteria,

displayed decreased ROS production and were suppressive to T-cell proliferation.

A unique subset of Low Density Neutrophils (LDN) has been implicated in the pathogenesis of

early stage autoimmune diseases such as Systemic Lupus Erythematous (SLE) and type 1

diabetes. SLE is an autoimmune disorder characterized by the presence of autoantibodies against

nuclear antigens. LDNs are reported to directly participate the pathogenesis of SLE as they are

more prone to death by NETosis resulting in the expulsion of host DNA material in the blood

stream. The self-DNA-antimicrobial protein complex has been shown to activate plasmacytoid

Dendritic Cells (pDCs) which in turn increase Interferon (IFN) – type I production. The INF-I

release initiates a perpetual positive feedback loop that beings with the activation of B

lymphocytes to produce autoantibodies against self-DNA and antimicrobial peptides released by

neutrophils and further activate neutrophils to release eve more NETs thus exasperating the auto-

immune disorder [110,122]. Similar to SLE, neutrophil activation of pDCs has also been

implicated in the pathogenesis of murine Type I Diabetes Mellitus [123].

2.4.2 Identification and Characterization and Cellular Subpopulations

A CD marker cluster is defined as a specific cohort of antibody clones that define a single cell

surface protein. The CD marker system allows for unambiguous identification of a specific cell

type or subpopulation based on their unique set of cell surface proteins [124]. However,

individual CD markers are often expressed in more than one cell type, which is why the

comprehensive and systematic characterization of specific CD markers are required for the

identification of unique cellular phenotypes and subpopulations. The analysis of a heterogeneous

cell population with the ultimate goal of discerning the phenotypes and subpopulations is called

Page 25: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

15

immunophenotyping [125]. Immunophenotyping requires the use of several CD markers and

unique gating strategies using a flow cytometer to accurately detect target populations [126]. The

identification of purified neutrophil populations and subpopulations relies determining cell size

and granularity, as well as on the expression profile of a number of CD markers. To date a

variety of CD markers including CD11b, CD14, CD16, CD62L, CD18, CD45 have been used for

the purification of neutrophils. Unfortunately, there has been little to no consistency in the use of

CD markers in the literature for the purification of neutrophils. For instance, Brooks et al. (2013)

used the combination of CD45mid (a marker of epithelial cells), CD14low (a marker of monocytes

and macrophages) and CD16+ (a marker of neutrophils) to select for neutrophils in fresh and

frozen sputum samples [127]. Fortunati et al. (2008) on the other hand, only used the

combination of CD45low and CD16high to select for neutrophils [128]. Conversely, in a

publication by Tak et al. (2015), circulatory granulocytes were isolated based on a combination

of forward-/side-scatter (FSC/SSC) and CD16+ while sputum neutrophils were identified as cells

that were CD11b+ (to exclude epithelial cells), 405/450mm-autflorescencelow (to exclude

macrophages) and CD16+ (to exclude eosinophils) [129]. Here we reviewed only three varying

examples of CD markers used to selectively isolate neutrophils from a heterogeneous population

using flow cytometry. To overcome this shortcoming and discrepancy in the literature, a

comprehensive High-Throughput Screening (HTS) assay was carried out to identify unique

neutrophil CD markers that are ubiquitously expressed in circulatory and tissue neutrophil

regardless of activation status. This project identified CD11b, CD66b, and CD16 as three

markers that are consistently expressed on neutrophils independent of the cell location, level of

activation and disease state. Cell sorting against CD11b, CD16 and CD66b allowed for the

enrichment of mature neutrophils, yielding neutrophil populations with up to 99% purity. These

findings suggest an ideal surface marker set for isolating mature neutrophils from humans.

In other publications, authors resort to using density gradients and centrifugation in varying

configurations to isolate specific cell populations. Density centrifugation relies on layering cells

with varying inherent densities onto a medium with a range of banding densities. Cells are then

spun at varying speeds such that cells will localize to a point where their density precisely

coincides with the density of the surrounding solution. Commonly used mediums in density

centrifugation include Percoll, Ficoll and Histopaque [130]. These media are generally used due

Page 26: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

16

to their biocompatibility and densities that are ideal for isolation of basophils, 1.080 g/mL

(1.075–1.081 g/mL); eosinophils, 1.088 g/mL (1.085–1.100 g/mL); neutrophils, 1.090 g/mL

(1.080–1.099 g/mL); lymphocytes, 1.077 (1.066–1.077 g/mL); and monocytes, 1.065 g/mL

(1.059–1.068 g/mL). Although technically simple, density centrifugation does have major

limitations. A study by Kuijpers et al. (1991) showed that expression of neutrophil surface

molecules is influenced by density centrifugation purification procedures. Centrifugation of

isolated circulatory neutrophils resulted in activation demonstrated by the increase of surface

CD10. Also, surface expression of several antigens that were expressed on circulating

neutrophils increased significantly after density-gradient centrifugation. The isolation method

caused increased expression of CD13, CD16, CD18, CD45, and CD67 but did not alter CD32

(FcRII), CD54 (ICAM-I), CD58 (LFA-3), Leu-8 and Human Leukocyte Antigen (HLA) class I

antigen expression [131].

2.5 Oral Neutrophils as a Unique Population of Neutrophils

Neutrophils are the most common circulatory leukocytes, yet relatively little is known about the

functional and biological diversity of these important cells as they migrate out of the circulation

and into peripheral organs. This can partly be attributed to the difficulty of collecting and

harvesting pure population from peripheral tissues. The gastrointestinal (GI) system is an

excellent model system to study tissue neutrophils in the context of inflammation and

inflammatory resolution [132]. Neutrophil have been shown to play a unique role in mediating

mucosal inflammation in the GI tract [133]. For instance, neutrophil depletion worsens the

inflammatory bowel disease symptoms in mice suggesting that neutrophils play a protective role

in GI mucosal tract [134][132]. Unfortunately, harvesting, collecting and analyzing human gut

neutrophils is inherently challenging and invasive. The oral cavity on the other hand, a terminal

end point of the GI system, is an accessible and abundant source of tissue neutrophils. Much like

GI mucosal neutrophils, oral tissue neutrophils play a critical surveillance role to help maintain

local equilibrium.

Neutrophils are the predominant the leukocyte in the oral cavity. Each hour, 2x106 neutrophils

are recruited out of the circulation and into periodontal connective tissues towards the gingival

sulcus where they are eventually washed out into the oral cavity with the Gingival Crevicular

Page 27: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

17

Fluid (GCF). Nearly all of the leukocytes that enter the oral cavity remain functional for many

hours [135] [136][137]. Once in the oral cavity, neutrophils can be readily harvested through

sequential oral saline rinses, with two minute rests to allow for steady-state recruitment of oral

neutrophils from the circulation. Oral rinses can then be pooled and neutrophils selectively

isolated through nylon mesh filtration [5,138]. Oral neutrophil counts in saliva are positively

correlated with the degree of inflammation in the oral cavity. Therefore, just as circulatory PMN

counts have been used a proxy for the determination of systemic health, oral neutrophil counts

can be used to assess the degree of periodontal disease and clinical inflammation and tissue

destruction in the oral [8]. That said, oral neutrophils do represent a unique neutrophil population

that does not always correlated or mimic circulatory neutrophils [139]. Oral neutrophils are

constantly recruited towards a healthy gingival sulcus in response to host and bacterial

inflammatory signals. Neutrophils are recruited to the oral cavity in germ free mice that lack

commensal oral bacteria suggesting that junctional epithelial cells alone are capable of

promoting neutrophil chemotaxis [140]. However, neutrophil recruitment to the oral cavity by

the healthy junction epithelial cells that are in direct contact with commensal microbiota is

significantly increased through the expression of an IL-8 gradient [141]. Again, significantly

more neutrophils are recruited oral cavity in the presence of periodontal disease [138]. While

traversing through the gingival sulcus, the neutrophil enriched GCF protects the dental junctional

epithelium from invasion and apical migration sub-gingival bacterial biofilm [142,143]. This is

supported by the observation that salivary leukocytes counts are lowest in completely edentulous

patients, intermediate in dentate patient lacking periodontal inflammation and highest in dentate

patients with clinical gingivitis [144]. Neutrophils play a protective role in the oral cavity

through phagocytosis, generation of ROS and degranulation. That said, neutrophils play a

doubled edged sword in oral cavity, particularly in the pathogenesis of periodontal disease. On

one hand, the absence of neutrophil in the periodontal tissues results in aggressive and highly

destructive forms of periodontal disease [145]. Conversely, excessive neutrophil recruitment and

hyperactive neutrophil response in periodontal tissues is directly correlated to disease severity

and pathogenesis [6,138][146].

Unfortunately, little effort has been devoted towards characterizing oral neutrophils in health

with the majority of published literature on oral neutrophils being aimed at characterizing the

Page 28: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

18

function and phenotypes of oral neutrophils in patients with chronic inflammation, typically

chronic periodontitis. Lakschevitz et al. (2013) examined the gene expression profile of blood

and oral neutrophils of healthy patients and those with chronic periodontitis. This study

demonstrated that there is a major gene expression change in oral neutrophils as they traverse out

of circulation and into the tissues regardless of oral disease status. There was a six-fold increase

in the degree of transcriptome change in oral neutrophils of patients with chronic periodontitis

compared to periodontally healthy patients. The gene profile of oral neutrophil in periodontitis is

characterized up-regulation in genes in the TLR and other inflammation pathways such as IL-1

and Chemokine Ligand (CCL) 3. In addition, alterations in genes regulating apoptosis were

noted, which is corroborated by the pro-inflammatory nature and prolonged longevity of oral

neutrophils in patients with choric periodontitis [5,147]. Oral neutrophils isolated from

periodontally healthy patients also display reduced phagocytic activity but similar levels of

intracellular bacterial killing when compared to circulatory neutrophils. Compared circulatory

neutrophils, oral neutrophils isolated from patients with chronic periodontitis were characterized

by enhanced hydrogen peroxide and ROS generation, greater potential for NETosis, enhanced in

chemotaxis, enhanced expression of CCL3 and IL-1 [5,148]. Taken together these findings are

reflective of the need for neutrophils to arrive at the site of inflammation and to carry out its

physiological roles.

Blood neutrophils isolated from patients with refractory chronic periodontitis, which does not

respond to conventional therapy, display higher levels of ROS production compared to blood

neutrophils of periodontally healthy patients [2]. These findings were induced by the induction of

ROS using PMA, a physiologically irrelevant yet useful laboratory tool. PMA directly activates

intracellular Protein Kinase C (PKC) independent of any membrane receptor ligands [2]. Other

studies, which induced ROS through the Fc receptor pathway, showed that CP peripheral

neutrophils produced more ROS compared to healthy blood neutrophils suggesting that CP

neutrophils have increased ore more responsive [146]. The capacity of blood neutrophil to

produce ROS is less critical than the ROS production capacity of tissue neutrophils. When

examining the ROS production of tissue neutrophils, studies have shown that oral neutrophils

have elevated baseline ROS production levels when compared to circulatory blood neutrophils

and that patients with more severe disease were characterized by oral neutrophils that produced

Page 29: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

19

the greatest levels ROS [6]. Concurrently, antioxidant (AO) levels in whole saliva of patients

with CP is significantly lower than in whole saliva of periodontally healthy patients [102].

Nuclear Factor Erythroid 2-Realted Factor (Nrf2) regulates the gene transcription of a group of

AO in response oxidative stress. Nrf2 binds to the promoter region of AO enzymes including

catalase and superoxide dismutase among others [149]. The Nrf2 pathway is down-regulated in

oral neutrophils of patients with severe CP compared to periodontally healthy patients. Ligature

induced periodontitis model in Nrf-/- null nice also displayed increased oxidative damage. The

Nrf2 pathway is key in the regulation and expression of cytoprotective factors (e.g. catalase,

superoxide dismutase), cell survival and tissue production during oxidative stress [7]. The

simultaneous down-regulation of the protective Nrf2 pathway in concert with increased

neutrophil recruitment to the oral cavity in CP provides a direct mechanism for tissue

destruction.

Page 30: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

20

Statement of the Problem

Neutrophil accumulation at sites of tissue injury and infection is a hallmark of acute

inflammation. Based on their classic functional role, neutrophils are viewed as pro-inflammatory

members of the innate immune system that seek and destroy invading pathogens and help

propagate the inflammatory response. In fact, improper and/or prolonged activation of

neutrophils can result in autoimmune and chronic inflammatory diseases characterized by

progressive tissue damage. However, the mere presence of neutrophils does not necessarily

imply disease. Research in neutrophil biology during the last three decades has elevated our

understanding of neutrophils. We now recognize neutrophils as sophisticated cells with

widespread roles in immunity and inflammation that extend far beyond the elimination of

pathogens. We further appreciate that in addition to their pro-inflammatory roles, neutrophils

also play a key role in establishing and maintaining tissue health and homeostasis. The paradox

of the destructive yet protective neutrophil is best exemplified in the oral cavity in the presence

and absence of chronic inflammation in the form of periodontitis. The etiology of periodontitis is

multifactorial and relies on the microbial, host and systemic factors. It is postulated that some

individuals may be less susceptible to periodontal disease due to an inherent ability to resist the

conversion of a symbiotic microbiota into a dysbiotic one. We have proposed that the missing

susceptibility link in periodontitis susceptibility lies within the neutrophil phenotypes of patients.

Therefore, we hypothesize that oral neutrophils isolated from the oral cavity of periodontally

healthy patients are functionally and phenotypically different than oral neutrophils isolated from

patients with chronic severe periodontitis. We further, propose that the quality and quantity of

oral neutrophils may be key in preventing or resisting periodontal breakdown in the face of a

microbial challenge. Using principles of immunophenotyping, flow cytometry and a unique

developed set of neutrophil specific cluster of differentiation markers we aim to show that in

healthy oral tissues, there exists an intermediary “para-inflammatory” immune state that allows

the host to respond to noxious agents without clinical signs of inflammation.

Page 31: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

21

Chapter 3 : Materials and Methods

3.1 Human Subjects

Participants were recruited from Toronto General Hospital’s Nephrology Center and University

of Toronto’s Graduate Periodontology Clinic. The study was approved by the University of

Toronto’s Research Ethic Board (#30044 & 29410) as well as the University Health Network

Research Ethic Board (#13-6896-AE). Signed informed consent was obtained from all

participants prior to inclusion. Following a preliminary complete dental examination, patients

with generalized severe CP [150] were recruited for the study. Volunteers with a healthy

periodontium and no history of disease, as defined by the absence of loss of clinical attachment,

were recruited as controls. Participants were excluded based on the following exclusion criteria:

1) pregnancy, 2) antibiotic or surgical periodontal therapy within the last six months 3) complete

edentulism and 4) other active oral disease including caries, endodontic lesions and

mucocutaneous diseases. After sample extraction, periodontal clinical parameters including

bleeding on probing (BOP), probing depth (PD), recession and clinical attachment level (CAL)

were measured at six sites per tooth. The degree of inflammation was determined by percentage

of sites that displayed bleeding upon probing [151]. A site was considered to have “active

disease” if it displayed bleeding on probing in combination with a probing depth of greater than

or equal to 5mm [152].

3.2 Sample Collection and Processing

Blood and oral samples were obtained as previously described [5]. Blood samples were drawn

into a vacutainer containing 0.1 volumes of sodium citrate as anticoagulant. With the exception

of electron microscopy and ROS assays, freshly drawn whole blood and oral rinse samples were

immediately fixed with 1.6% paraformaldehyde (PFA) for 15 minutes at 4oC, which we found

preserved native surface expression of CD markers without altering antigenicity. Erythrocytes

were eliminated by isotonic lysis. Oral samples were pelleted at 690 x g and resuspended in 10

ml of cold Phosphate Buffered Saline (PBS) and epithelial cells were removed by filtration as

previously described [153]. Cell sorting based on CD18 expression was performed to confirm

>98% purity of oral neutrophil populations.

Page 32: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

22

3.3 Electron Microscopy

Fresh oral neutrophils were prepared as above, without PFA fixation. Sample preparation and

electron microscopy were performed as previously described [154]. Briefly, cells were pelleted

and fixed with Karnovsky’s style fixative (3.2% paraformaldehyde + 2.5% glutaraldehyde in a

0.1M Sorensen’s phosphate, pH 6.8 - 7.0) for one hour. Fresh fixative was replaced and samples

were left over night at 4oC. After removal of the fixative the pellets were washed with Sorensen’s

phosphate buffer (A mixture of 0.1 M monobasic and 0.1 M dibasic sodium phosphate salts in a

ratio to give the required pH) and then post-fixed with 1% osmium tetroxide for one hour.

Following another wash, the pellets were dehydrated using a graded series of ethanol/distilled

water at 30%, 50%, 70%, 95% and 100% ethanol. The pellets were then washed with the

transitional solvent propylene oxide. Infiltration was performed using Epon Araldite resin,

prepared as described previously [155], using a graded series of Epon Araldite (E/A) and

propylene oxide (PO) at 1:1 E/A and PO for half an hour, 3:1 E/A and PO for two hours, and

100% E/A overnight. Another infiltration using 100% E/A was performed the next day for

another two hours. The samples were then placed in polyethylene BEEM capsules with fresh

E/A, and placed in an oven for polymerization for 48 hours at 60°C. After complete

polymerization, the solid resin blocks containing the samples were sectioned on a Reichert

Ultracut E microtome to 60 to 90 nm thickness and collected on 200 mesh copper grids. The

sections were counter stained using saturated uranyl acetate for 15-20 minutes, rinsed in distilled

water, followed by Reynold’s lead citrate for 15-20 minutes and rinsed again in distilled water.

The sections were examined and photographed in a Hitachi H7000 transmission electron

microscope at an accelerating voltage of 75Kv. Image analysis was performed using ImageJ

software.

3.4 Histopaque Density Centrifugation

Discontinuous density gradients were generated by carefully layering equal volumes of

1.077g/ml and 1.119g/ml Histopaque centrifugation medium (Sigma). Whole blood and saliva

rinse samples were layered onto the gradient and centrifuged at 1000 x g for 35 minutes. High-

density (HD) cells were collected from the 1.077-1.119 interface, while low-density (LD) cells

were collected from the plasma-1.077 interface. Contaminating erythrocytes were eliminated via

hypotonic lysis.

Page 33: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

23

3.5 Multicolor Flow Cytometry

Whole blood leukocytes and oral neutrophils (5 x 105) were resuspended in 50 µl of

Fluorescence-Activated Cell Sorting (FACS) buffer and labeled with two separate panels of

antibodies as detailed in Table 1. The markers were classified into four categories based on

function: degranulation/activation markers (CD10, CD63, CD64 and CD66a), immunoregulation

markers (CD16 and CD170), adhesion markers (CD11b, CD18 and CD177) and complement

regulators (CD55). Cells were labeled for 30 minutes on ice in the dark and washed three times

with FACS buffer. At least 2 x 104 gated events were acquired using an LSR Fortessa (BD

Biosciences) flow cytometer. For each CD marker, appropriate fluorescently tagged isotype

control antibodies were used to determine autofluorescent signals, which were subtracted from

Mean Fluorescence Intensities (MFI). Flow cytometer channel voltages were calibrated manually

using rainbow beads in order to normalize sample acquisition on different days. Compensation

was performed with single stained OneComp eBeads (eBioscience). Gating was performed as

described in (Figure 3A and B). Doublets were excluded by SSC-W x SSC-H. Data were

analyzed using FlowJo (vX) software. Samples from one healthy volunteer were run on multiple

occasions to confirm reproducibility on different days. Also, samples stained in duplicate on a

given day were found to yield identical results.

3.6 ROS Assay

ROS assays were performed essentially as described [6]. Unfixed oral neutrophils were prepared

and incubated for 20 minutes at 37°C in Hanks-/- containing Dihydrorhodamine (DHR) at a final

concentration of 2 µM, and stimulated for an additional 15 minutes with Phorbol 12-Myristate

13-Acetate (PMA) at a final concentration of 200 nM, or left unstimulated. The cells were placed

on ice and labeled with CD18-BV421. Flow cytometry, gating and analysis were performed as

described above. The use of DHR with flow cytometry to measure ROS production has

previously been validated and has been proven to be a fast, reliable and easy method to evaluate

ROS production [2].

Page 34: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

24

3.7 NET Assay

Preparation of fixed, purified oral neutrophils was performed as above. Flow cytometric analysis

of NET formation was performed as described previously [156]. Oral neutrophils were labeled

sequentially with primary anti-Histone H3 (Citruline R2 + R8 + R17, Abcam) and secondary

goat anti-rabbit-AF488 (Abcam) antibodies, and then labelled with MPO-PE (Clone: 2C7, Acris)

and CD18-BV421. Each incubation was for 30 minutes, followed by one wash with FACS

buffer. Flow cytometry, gating and analysis were performed as described above.

3.8 Statistical Analysis

One-way ANOVA was performed with a post-hoc Tukey’s test for pairwise comparisons.

Clinical parameters were compared using Student’s t-test. P ≤ 0.05 were considered statistically

significant. Statistical analysis was performed using GraphPad software

Page 35: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

25

Chapter 4 : Results

4.1 Clinical Description of Periodontal Status

Seventeen CP patients and 11 healthy controls were recruited in order to characterize blood and

oral neutrophil phenotypes. On average, patients with CP had 3.1 fewer teeth (24.9 ± 4.2) than

healthy controls (28 ± 1.7, p = 0.05), with BOP occurring at 62% of sites in CP patients

compared to 8.6% of sites in healthy controls (p < 0.001). In patients with periodontal disease,

25.5% of all sites were deemed to be undergoing active inflammation-mediated periodontal

tissue breakdown, while active disease (pocket depth ≥ 5 with BOP) was virtually absent (0.1%)

in healthy controls (p < 0.001). Consistent with previous observations [138], patients with CP

had an elevated oral inflammatory cell load as demonstrated by a five-fold increase in oral

neutrophil counts compared to healthy controls (p = 4 x 10-4). There were no significant

differences between the two groups with respect to age, age range or gender (Table 2).

4.2 CP Oral Neutrophils Are More Degranulated and Have Increased

Phagocytosis Compared to Healthy Oral Neutrophils

In order to characterize functional differences between blood and oral neutrophils in health and

disease, we evaluated granule content and phagocytosis in CP and healthy oral neutrophils by

electron microscopy (Figure 4). The mean number of granules per µm2 of cytoplasm was lower

in oral neutrophils compared to blood neutrophils (Figure 4B), and granule content was further

reduced in oral neutrophils from CP patients compared to healthy controls. There were no

differences in granule content between blood neutrophils of healthy and CP patients.

Phagosomes were commonly observed in oral neutrophils, but not in blood neutrophils (Figure

4C). Oral neutrophils from chronic periodontal disease patients contained, on average, more

early and late phagosomes than oral neutrophils from healthy controls. Elevated degranulation

and phagocytosis by CP oral neutrophils confirms that these are in a heightened inflammatory

state compared to healthy oral neutrophils.

Page 36: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

26

4.3 Flow Cytometric Gating Strategy

Oral rinse samples contain neutrophils, epithelial cells, bacteria and debris. Epithelial cells were

removed by nylon mesh filtration, and we found that gating on CD18+ve was sufficient to exclude

debris (Figure 3A). Cytological analysis of sorted cells confirmed the purity of neutrophils in

CD18+ve populations, while CD18-ve populations contained debris but no cells (Figure 3A).

Neutrophils in whole blood were gated based on high CD16 expression in combination with SSC

(Figure 3B). Although a subset of circulating monocytes are known to be CD16+ve [157],

analysis of high and low density blood fractions confirmed that these monocytes had lower SSC-

A and lower CD16 expression than blood neutrophils and were excluded by our gating strategy

(results not shown).

4.4 Characterization of Oral Neutrophil Populations in Health and CP

We performed high-resolution multicolor flow cytometry to analyze CD marker signatures of

blood and oral neutrophils, and to identify and immunophenotype neutrophil subsets in health

and CP. Two panels of antibodies were developed based on an HTS assay [44] and literature

confirming the relevance of specific neutrophil CD markers in human inflammatory conditions

(Table 1). All of the markers that were selected had significantly altered expression on oral

neutrophils compared to blood neutrophils in the HTS screen. Analysis of FSC x SSC, which are

generally equivalent to cell size and granularity, indicated that oral neutrophils were shifted

towards the lower left quadrant compared to blood neutrophils (Figure 5). In healthy oral

neutrophil samples, we identified two populations with distinct epicenters based on FSC x SSC

(Figure 5A and C), while only one population was observed in CP (Figure 5B and D). The two

oral neutrophil populations in health, which occurred in roughly equal proportions, were: a

population with similar FSC-A x SSC-A properties to blood neutrophils (P1, 45.2% ± 3.2), and a

population that was shifted to the lower left quadrant of the scatterplots (P2, 50.3% ± 3.2)

(Figure 5A). The CP oral neutrophil population was consistently observed in the lower left

quadrant of scatterplots (P3) (Figure 5B). The mean geometric MFI of each CD marker was

determined for blood neutrophils and for each oral neutrophil population (Figure 6 and Table

3). There was no difference in expression of CD markers between blood neutrophils of healthy

volunteers and CP patients. All of the CD markers in our panels had elevated expression on each

Page 37: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

27

of the oral neutrophil populations compared to blood, with the exception of CD16, which had

reduced expression. By comparing oral neutrophil populations, we found that seven CD markers;

CD10, CD11b, CD18, CD55, CD63, CD64 and CD66, had significantly elevated expression on

CP oral neutrophils compared to either of the oral neutrophil populations in health. Furthermore,

CD16 and CD170 had elevated expression on the CP oral neutrophils compared to the P2 healthy

oral neutrophils, but similar expression to the P1 healthy oral neutrophils. Comparing the two

healthy oral neutrophil populations, we found significant differences in expression of four CD

markers. Expression of CD55 and CD63 were elevated, while the expression of CD16 and

CD170 were reduced on the P2 population compared to the P1 population. Retrospectively, we

found that gating based on expression of CD16 and CD170 markers was sufficient to distinguish

between the two healthy oral neutrophil populations (Figure 7). Based on our observation of

higher degranulation, phagocytosis and expression of neutrophil markers of activation by CP oral

neutrophils, this population was designated as pro-inflammatory (Pro), while the two healthy oral

neutrophil populations, based on the para-inflammation hypothesis, were called Para1 (FSC-

Ahigh/SSC-Ahigh) and Para2 (FSC-Alow/SSC-Alow).

4.5 Pro-inflammatory Neutrophils Have Elevated ROS Production and

NET Formation

To assess the functionality of oral neutrophil populations, we examined baseline and PMA

stimulated generation of ROS by flow cytometry. By gating on the two healthy oral neutrophil

populations (Para1 and Para2), we found that the Para2 population produced more ROS than the

Para1 population at baseline, and both populations showed increased ROS production after

stimulation with PMA (Figure 8A). Pro-inflammatory neutrophils produced more ROS than the

para-inflammatory oral neutrophils. However, in contrast to the Para1 and Para2 populations,

PMA stimulation did not induce a significant increase in ROS production by the CP neutrophils,

suggesting that they have exhausted their potential for ROS production, while para-inflammatory

neutrophils retain their potential for further stimulation (Figure 8A). A flow cytometric NETosis

assay, which detects myeloperoxidase (MPO) and histone citrullination on the cell surface, was

used to detect neutrophil extracellular trap (NET) formation in oral neutrophil populations. We

found that Pro oral neutrophils from CP patients showed high levels of NET formation,

compared to the Para cells from healthy patients (Figure 8B).

Page 38: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

28

Chapter 5 : Discussion

The present study is the first to characterize oral neutrophil subsets specific to health and chronic

inflammation of the oral mucosa. This reports indicates that oral neutrophils from CP patients are

in a pro-inflammatory activation state compared to healthy oral neutrophils, as determined by

elevated degranulation, phagocytosis, ROS production, NETosis and a characteristic signature of

cell surface markers of activation. We thus demonstrate an intermediate or para-inflammatory

state of oral neutrophils in health. Neutrophils are important primary innate immune responders

that migrate from the circulation to sites of inflammation in the tissue. Neutrophils contribute to

periodontal tissue homeostasis through microbial surveillance and direct modulation of the host

tissue response [158], however pathological neutrophil responses can result in tissue damage

[95,159,160]. While multiple circulating neutrophil subtypes are known [17,19,114,161], to our

knowledge, diversity of neutrophil populations in tissue has not been studied, nor is there an

understanding of how neutrophils maintain tissue homeostasis in the presence of commensal

bacteria. CD markers and their surface expression levels can be used for three main purposes: 1)

To label/define a specific population of interest, 2) as markers of functionality or 3) to gauge the

state of activation of a particular function [162,163]. Here we identified separate neutrophil

populations with unique phenotypes in health and disease using custom CD marker panels, and

compared the functional activity of these populations. We found CD markers indicative of cell

activation to be expressed at much higher levels on oral neutrophils in CP compared to healthy

oral neutrophils, constituting a multiparameter CD marker signature that can differentiate

between para- and pro-inflammatory tissue neutrophil populations, based on activation state. The

markers that were up-regulated on CP oral neutrophils fall into three categories: 1) markers of

activation/degranulation; CD10 [131], CD63 [164,165], CD64 [166,167] and CD66a [168], 2)

adhesion receptors: CD11b and CD18, and 3) complement inhibitor CD55 [169]. The pro-

inflammatory phenotype of CP oral neutrophils was confirmed by elevated degranulation,

phagocytosis, ROS production and NETosis.

Interestingly, we observed two different populations of oral neutrophils in health. The Para1 and

Para2 populations differed based on their size and granularity profile, expression of specific CD

markers, production of ROS and NET formation. The Para1 oral neutrophil phenotype, which is

present only in health, has a FSC and SSC profile that is similar to naïve blood neutrophils, and

Page 39: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

29

these cells are in a lower state of activation relative to the Para2 oral neutrophils, based on the

relative expression levels of four CD markers. Compared to the Para1 population, Para2 oral

neutrophils had elevated expression of two markers of activation (CD55 and CD63) and reduced

expression of the inhibitory receptor, CD170 [170], and the low affinity Fc-receptor, CD16. A

drop in CD16 is likely a consequence of internalization as a result of phagocytosis, and therefore

is consistent with elevated phagocytic activity of the Para2 population compared to the Para1

population. Functionality, as determined by ROS production and NET formation, was also

reduced in the Para1-neutrophils compared to Para2-neutrophils. We surmise that the Para2

neutrophils may be the front line neutrophils within the healthy tissue that interact with the

commensal biofilm, or are coming from pockets with increased biofilm load, resulting in an

increased activation state that is still below the maximal activation state of the Pro-inflammatory

neutrophil population observed in CP. Similarly, Para1 neutrophils, which also show

upregulation of activation markers compared to blood, but have restrained activation compared

to Para2 neutrophils, could be oral neutrophils that are in a primed state.

In contrast to health, there is only one population of CP oral neutrophils, which have a similar

FSC/SSC profile to Para2-neutrophils, but are more activated than either of the Para-

inflammatory populations based on ROS production, NET formation and expression of CD

markers of activation. This suggests that periodontal tissue neutrophils lie on a spectrum of

increasing activation; Para1 < Para2 < Pro. Based on the fact that the Para1 and Para2

populations occur in equal proportions, and were absent in CP, it is possible that the balance

between the two populations contributes to the maintenance of tissue health in the presence of

commensal microorganisms. Another possibility is that Para2 neutrophils come from sites of

undetected subclinical inflammation in the mouth. While CD16 and CD170 expression are

reduced in Para2 populations compared to Para1, levels of these two markers remain high on

pro-inflammatory neutrophils in CP. It is unclear why CD16 and CD170 expression remain high

on the CP oral neutrophils, since this would predict that these cells are not fully activated. It is

possible that periodontal pathogens may be involved in the up-regulation of these markers as part

of their immune-modulatory actions, causing suppression of certain aspects of neutrophil

function.

Reduced SSC of Para2 and Pro neutrophils, relative to the Para1 population, are consistent with

increased degranulation in these populations, however, nuclear morphological changes including

Page 40: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

30

those associated with NETosis might also alter the spectral properties of the cells. Upregulation

of CD markers associated with granules confirms that healthy and CP oral neutrophil populations

have undergone degranulation relative to blood neutrophils [91]. Furthermore, a relative increase

in degranulation markers on oral neutrophils in CP, compared to healthy oral neutrophils, is

consistent with increased degranulation observed by electron microscopy, and suggests that

reduced SSC is at least partly due to increased degranulation in these populations.

In CP there is a shift from two para-inflammatory populations to one pro-inflammatory

neutrophil phenotype in response to the dysbiotic microenvironment instigated by pathogenic

microbes. This conversion of para-inflammatory neutrophils into pro-inflammatory neutrophils is

likely to be a key aspect of pathogenesis in periodontal disease. Further research will be

necessary to elucidate the functional interplay between the two para-inflammatory subtypes.

Furthermore, experimental models of gingivitis will be useful to better understand the transition

from Para1 to Para2 to Pro neutrophils.

Page 41: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

31

Conclusion and Future Directions

Inflammation is the physiological response to a noxious stimulus. Chronic inflammation can be

thought of as the extreme end of a spectrum of tissue states that ranges from steady state

homeostasis, to protective para-inflammation and finally to acute or choric inflammation. In the

healthy oral cavity there is a constant low-grade stress caused by repeated physical trauma and

presence of commensal microbiome. This is countered by a constant influx of neutrophils that

help establish and maintain tissue homoeostasis. Based on the outcomes of this study, we can

conclude that the neutrophils recruited to the healthy oral cavity are in an intermediate or Para-

inflammatory state and have not been activated to their full potential. Neutrophils are classically

thought to be in either a resting or activated state, however we have demonstrated that they

actually exist in four states: a resting basal state in blood, two para-inflammatory states in the

healthy oral cavity and a fully activated pro-inflammatory state in the diseased oral cavity. Oral

neutrophils from CP patients are in a Pro-inflammatory state when compared to healthy oral

neutrophils (Para-1 and Para-2), based on elevated degranulation, phagocytosis, ROS & NET

production and CD marker expression profile (Figure 9A). Para-1 oral neutrophils have a flow

cytometry profile that is similar to naïve blood neutrophils and show upregulation of activation

markers compared to blood neutrophils but have restrained activation compared to Para- 2

neutrophils. Therefore, Para-1 neutrophils represent oral neutrophils that have recently migrated

out of the circulation into the tissues and are in a primed state. Para-2 neutrophils have an

intermediary phenotypic and functional state between Para-1 and Pro-inflammatory neutrophils.

Para-2 oral neutrophils have a flow cytometry profile that resembles CP oral neutrophils. On a

functional level they produce intermediate level of NETs and ROS but can be further stimulated

towards Pro-inflammatory neutrophils. Para-2 neutrophils have elevated expression of some

activation markers (CD55 and CD63) and reduced expression of the inhibitory receptor CD170

and the low-affinity Fc receptor, CD16 relative to Para-1 neutrophils. These differences are

consistent with elevated phagocytic activity of the Para-2 population. Para-2 neutrophils likely

represent front-line neutrophils within the healthy tissue that interact with the commensal biofilm

resulting in an increased activation state that is still below the maximal activation state of the

pro-inflammatory neutrophil population observed in CP. Para-2 inflammatory neutrophil subsets

arise in response to localized inflammatory cues within the tissue and may serve to protect

tissues from host mediated tissue damage. The host protective role of para-inflammatory

Page 42: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

32

neutrophils has yet to be clinically proven and requires further investigation. The current study

has made significant advancement in our understanding of oral neutrophils. However, there are

certain limitations that need to be addressed. First and foremost, the findings of this study need to

be repeated and validated using a large sample size of patients. Secondly, the presence and

function of Para-inflammatory neutrophils in patients with more intermediate levels of

periodontal disease as well as patient is gingivitis should be investigated. This study only focused

on the extreme ends of periodontal health to disease to facilitate the identification of key

differences between oral neutrophil phenotypes in health and disease. Lastly, the possible

presence of role of Para-inflammatory neutrophils should be investigated in other chronic

inflammatory conditions such as diabetes mellitus, inflammatory bowel disease, ulcerative colitis

and rheumatoid arthritis.

Page 43: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

33

Figure Legend

Figure 1. Neutrophils in the Maintenance of Periodontal Health and Pathogenesis of

Chronic Periodontitis. Blood neutrophils are constitutively recruited to the periodontal tissue

and transition into the mouth. In periodontal health, there is a steady state recruitment of

neutrophils in the healthy oral cavity that is capable of co-existing with the commensal flora

without initiating frank inflammation and host tissue destruction (left panel). In CP, greater

number of neutrophils enter the gingival sulcus in response to pro-inflammatory mediators

generated by pathogenic bacteria and damaged host tissues. The resulting dysbiotic/pro-

inflammatory environment results in destruction of connective tissues in the periodontium, loss

of attachment and eventual tooth exfoliation (right panel).

Figure 2. Etiology and Pathogenesis of Chronic Periodontitis. The etiology of periodontitis is

multifactorial and relies on the microbial, host and systemic factors. At the microbial level,

periodontitis pathogenesis is driven by the quality and quantity of the biofilm. In the transition

from health to disease there is a micro-environmental shift that disrupts a healthy and

harmonious (“symbiotic”) biofilm into a pathogenic and destructive (“dysbiotic”) microbial

community that promotes periodontal breakdown. Bacterial dysbiosis may be required to initiate

periodontitis but often fails to cause periodontitis. Behavioural (e.g. smoking), systemic (e.g.

diabetes) and genetic (e.g IL-1B polymorphism) risk factors can account for half of the variation

in periodontal disease susceptibility.

Figure 3. Multicolor Flow Cytometry Gating Strategy for Blood and Oral Neutrophils.

Scatterplots of representative blood and oral neutrophil samples from a healthy individual are

shown to demonstrate the gating strategy used for multicolor flow cytometry. Doublets were

excluded based on SSC-H x SSC-W. (A) Oral neutrophils were gated based on expression of

CD18 to exclude debris. Cytospins of CD18+ve and CD18-ve sorted cells were analyzed by Diff-

Quik staining. Unfiltered oral cells are shown for reference. Bars are 10 µm. (B) Neutrophils in

whole blood were gated using CD16hi/SSC-A.

Figure 4. Oral Neutrophils Have Elevated Phagocytosis and Greater Degranulation in CP.

Blood and oral neutrophils were pelleted, fixed and imaged by electron microscopy. (A)

Page 44: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

34

Representative images are shown. Arrowheads indicate granules and arrows indicate

phagosomes. Bar is 2 µm. Granule number normalized to cytoplasmic area (B) and phagosomes

per cell (C) were determined using ImageJ software. At least 25 cells were analyzed from paired

blood and oral neutrophils of controls (n = 5) and periodontal disease patients (n = 5). Significant

differences were determined by ANOVA with a post hoc t-test. *<0.05.

Figure 5. Identification of Para-inflammatory Neutrophils in Oral Health. Representative

contour plots of blood and oral neutrophils from a healthy (A) volunteer and a CP (B) patient.

Two oral populations were observed in health, in roughly equal proportions, and were gated

based on FSC x SSC profiles. In CP, only one population of oral neutrophils was observed with

low FSC and low SSC. Gates for the three oral neutrophil populations (P1, P2 and P3) are

shown. Conversely, only a single Pro-inflammatory neutrophil population was identified in oral

samples of patients with CP. FSC x SSC profiles of five representative oral neutrophil samples

from health (C) and CP (D) are shown. Bi-modal populations in health were gated as P1 and P2,

as indicated. Percentages of total neutrophils are indicated.

Figure 6. Para- & Pro-inflammatory Oral Neutrophil Populations Are Defined by Unique

CD Marker Expression Profiles. Blood and oral neutrophils of healthy controls (n=11) and CP

patients (n=17) were analyzed by multicolour flow cytometry. (A) Representative histograms

expression on blood neutrophils (red), P1 oral neutrophils (green), P2 oral neutrophils (orange)

and P3 oral neutrophils (blue) are shown. FMO isotype controls are indicated for each CD

marker. (B) The geometric MFI for each CD marker is shown for the healthy (P1, P2) and CP

(P3) oral neutrophil populations. Mean geometric MFI ± SEM are shown. ANOVA with a post-

hoc t-test was performed to determine statistical significance (**P < 0.01).

Figure 7. CD Markers Can Be Used to Gate on Para1 and Para2 Oral Neutrophil

Populations. Oral neutrophil populations were analyzed based on CD16 x CD170 (A) and CD16

x CD55 (B). Contour plots for a representative healthy subject are shown.

Figure 8. Pro-inflammatory Oral Neutrophil Populations Have Elevated NET Formation

and Increased ROS Production Compared to Para-inflammatory Neutrophils. (A) Oral

neutrophils from healthy controls and CP patients were labelled with DHR and treated with PMA

(+PMA) or left untreated (-PMA) at 37°C for 15 minutes. Representative histograms showing

levels of the DHR oxidation product, rhodamine 123, on Para1, Para2 and Pro-inflammatory

Page 45: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

35

neutrophil populations, are shown. Fluorescence minus one (FMO) controls are indicated. At

least 2 x 104 events were acquired in three independent experiments. Bar graphs show basal

mean geometric MFI’s of rhodamine 123 signal and the fold-increase in MFI in response to

PMA stimulation for each oral neutrophil population. (* P < 0.05, n=3). (B) Fixed oral

neutrophils were labelled with H3Cit, MPO and CD18 antibodies and analyzed by flow

cytometry. Doublets were excluded and neutrophils were gated based on expression of CD18.

Representative histograms of H3Cit and MPO expression on Para1, Para2 and Pro-inflammatory

neutrophil populations, are shown. FMO controls are indicated. At least 2 x 104 events were

acquired in three independent experiments. Bar graphs show mean geometric MFI’s for H3Cit

and MPO for each oral neutrophil population ± SEM. (* P < 0.05, ** P < 0.01, n=7).

Figure 9. Model of Para- and Pro-Inflammatory Neutrophil Phenotypes in Health and

Disease. Para-inflammatory neutrophil subtypes are present in periodontal health while pro-

inflammatory neutrophils occur in CP. The two para-inflammatory populations in health differ

based on CD marker expression and functional activity. Para2 oral neutrophils produce more

ROS and NETs compared to Para1 populations. In addition to elevated neutrophil recruitment

during CP, Pro-inflammatory neutrophils are in a heightened inflammatory state based on high

expression of CD markers of activation, elevated production of ROS and more NET formation

compared to both para-inflammatory oral neutrophils.

Page 46: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

36

Tables and Figures

Figure 1. Neutrophils in the Maintenance of Periodontal Health and Pathogenesis

of Chronic Periodontitis.

Page 47: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

37

Figure 2. Etiology and Pathogenesis of Chronic Periodontitis

Page 48: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

38

Table 1. CD Marker Panels for Multicolor Flow Cytometry

CD

marker

Protein - Function Flour Supplier Clone

Panel 1

CD16 Fc-γRIII – Low affinity Fc-

receptor

PE Biolegend 3G8

CD11b αM-integrin – Adhesion,

complement receptor

PerCP-Cy5.5 Biolegend ICRF44

CD177 NB1 - Adhesion APC Abcam MEM-

166

CD64 Fc-γRI – High affinity Fc-receptor Alexa700 BD 10.1

CD10 Neprylisin – Inflammatory

metalloproteinase

APC-Cy7 Biolegend HI10a

CD18 2-integrin – Adhesion,

complement receptor

BV421 BD 6.7

Panel 2

CD55 DAF – Complement inhibitor FITC BD IA10

CD170 Siglec-5 – Inhibitory receptor,

adhesion

PE R&D 194128

CD63 Granulophysin – Degranulation PerCP-Cy5.5 Biolegend H5C6

CD66a Ceacam – Adhesion, degranulation APC eBioscience CD66a-

B1.1

CD16 Fc-γRIII – Low affinity Fc-

receptor

Alexa700 BD 3G8

CD11b αM-integrin – Adhesion,

complement receptor

APC-Cy7 Biolegend ICRF44

CD18 2-integrin – Adhesion,

complement receptor

BV421 BD 6.7

Page 49: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

39

A

FSC-A SSC-H CD18 FSC-A

SSC

-A

SSC

-W

SSC

-A

SSC

-A

Ora

l

Unfiltered Debris Neutrophils

Blo

od

B

FSC-A SSC-H CD16 FSC-A

SSC

-A

SSC

-W

SSC

-A

LDHD CD16+

SSC

-A

C

CD16

SSC

-A

Figure 3. Multicolor Flow Cytometry Gating Strategy for Blood and Oral

Neutrophils

Page 50: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

40

Table 2. Demographic and Periodontal Characteristics of Patients Cohorts

*Mean ± STDev †Neutrophil yield was per 20ml of saliva

Characteristic Healthy (n=11)

Diseased (n=17)

Intergroup P-value

Age (years)* 49 ± 18 57 ±13 0.16

Female (%) 9 (58%) 9 (41%) 0.23

Smoking 0.07

Never 10 9 -

Former smoker (years ago) 1 (10) 5 (15-50) -

Current smoker 0 3 -

Number of teeth* 28 ± 1.7 24.9 ± 4.2 0.04

Number of sites* 168 ± 10.2 150.± 25.5 0.04

% Sites with BOP* 8.6 ± 10.4 62. ± 18.5 1.06 x 10-8

Active disease (PD ≥ 5mm + BOP)

Number of sites with active disease* 0.2 ± 0.6 39.5 ± 26.4 8.63 x 10-5

Percent sites with active disease* 0.1 ± 0.4 25.5 ± 15.5 2.74 x 10-5

Neutrophil yield (x 106)*† 1.1 ± 0.6 5.2 ± 3.8 4 x 10-4

Page 51: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

41

Figure 4. Oral Neutrophils Have Elevated Phagocytosis and Greater Degranulation

in CP

Page 52: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

42

Figure 5. Identification of Para-inflammatory Neutrophils in Oral Health

SSC

SS

C

FSC

C

D

FSC

Page 53: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

43

Figure 6. Para- & Pro-inflammatory Oral Neutrophil Populations Are Defined by

Unique CD Marker Expression Profiles

Page 54: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

44

Page 55: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

45

Table 3. CD Marker Expression levels on Healthy and CP Neutrophils

Marker

BloodH

(n=11), Mean

geometric MFI

± SEM (x 103)

Para1

(n=11), Mean

geometric MFI ±

SEM (x 103)

Para2

(n=11), Mean

geometric MFI

± SEM (x 103)

BloodCP

(n=17), Mean

geometric MFI

± SEM (x 103)

Pro

(n=17), Mean

geometric MFI

± SEM (x 103)

CD10 0.12 ± 0.01 1.27 ± 0.17 1.26 ± 0.18 0.10 ± 0.01 2.06 ± 0.24

CD11b 0.18 ± 0.02 3.34 ± 0.78 3.42 ± 0.56 0.16 ± 0.01 6.70 ± 0.60

CD16 7.84 ± 0.29 4.92 ± 0.33 2.10 ± 0.18 7.13 ± 0.22 3.82 ± 0.45

CD18 1.72 ± 0.08 8.91 ± 0.62 8.54 ± 0.60 1.72 ± 0.11 13.06 ± 1.07

CD55 0.87 ± 0.05 3.13 ± 0.36 4.72 ± 0.70 0.84 ± 0.02 6.71 ± 0.65

CD63 0.37 ± 0.02 6.47 ± 0.98 9.14 ± 0.99 0.34 ± 0.02 19.66 ± 2.22

CD64 0.24 ± 0.01 0.61 ± 0.07 0.71 ± 0.09 0.23 ± 0.01 1.12 ± 0.10

CD66a 0.78 ± 0.04 9.00 ± 0.62 9.04 ± 0.70 0.73 ± 0.02 14.28 ± 1.16

CD170 2.69 ± 0.25 24.32 ± 1.91 12.54 ± 1.09 2.41 ± 0.12 24.01 ± 3.25

CD177 3.28 ± 0.26 7.19 ± 1.17 5.36 ± 0.91 3.13 ± 0.29 9.81 ± 2.27

Page 56: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

46

Figure 7. CD Markers Can Be Used to Gate on Para1 and Para2 Oral Neutrophil

Populations

A

B

Page 57: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

47

Figure 8. Pro-inflammatory Oral Neutrophil Populations Have Elevated NET

Formation and Increased ROS Production Compared to Para-inflammatory

Neutrophils

Page 58: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

48

Figure 9. Model of Para- and Pro-Inflammatory Neutrophil Phenotypes in Health

and Disease

A

Page 59: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

49

Contributions to the Thesis and Manuscript

Siavash Hassanpour is solely responsible for the entire content of this thesis.

The contents of this thesis is based on the manuscript entitled “Distinct Oral Neutrophils Subsets

Define Health and Periodontal Disease States” [43]. The contributions to the manuscript were as

follows:

Siavash Hassanpour contributed to the conception of the project, the design of the experiments,

sample preparation, data analysis, interpretation of the data and preparation of the manuscript.

Siavash Hassanpour was solely responsible for the screening and recruitment of all patients, as

well as sample and patient data collection.

Noah Fine contributed to the conception of the project, design of the experiments, sample

preparation, data analysis, interpretation of the data and preparation of the manuscript. Noah Fine

was solely responsible for all flow cytometry based experiments.

Alon Borensetin contributed by performing the electron microscopy experiments.

Page 60: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

50

References

[1] Nauseef WM, Borregaard N. Neutrophils at work. Nat Immunol 2014;15:602–611.

[2] Johnstone AM, Koh A, Goldberg MB, Glogauer M. A hyperactive neutrophil phenotype in

patients with refractory periodontitis. J Periodontol 2007;78:1788–1794.

[3] Eke PI, Dye BA, Wei L, Slade GD, Thornton-Evans GO, Borgnakke WS, et al. Update on

Prevalence of Periodontitis in Adults in the United States: NHANES 2009 to 2012. J Periodontol

2015;86:611–622.

[4] Socransky SS, Haffajee AD. Evidence of bacterial etiology: a historical perspective.

Periodontol 2000 1994;5:7–25.

[5] Lakschevitz FS, Visser MB, Sun C. Neutrophil transcriptional profile changes during transit

from bone marrow to sites of inflammation. Cell Mol Immunol 2014;12:53–65.

[6] Aboodi GM, Goldberg MB, Glogauer M. Refractory periodontitis population characterized

by a hyperactive oral neutrophil phenotype. J Periodontol 2011;82:726–733.

[7] Sima C, Aboodi G, Lakschevitz F, Chunxiang S, Goldberg M, Glogauer M. Nuclear Factor

Erythroid 2-Related Factor 2 Down-Regulation in Oral Neutrophils Is Associated with

Periodontal Oxidative Damage and Severe Chronic Periodontitis. The American Journal of

Pathology 2016;186:1417–1426.

[8] Bender JS, Thang H, Glogauer M. Novel rinse assay for the quantification of oral neutrophils

and the monitoring of chronic periodontal disease. J Periodontal Res 2006;41:214–220.

[9] Page RC, Schroeder HE. Pathogenesis of inflammatory periodontal disease. A summary of

current work. Lab Invest 1976;34:235–249.

[10] Raeste AM, Tapanila T, Tupakka R. Leukocyte migration into the healthy dentuious mouth

A study in children, adolescents and adults. J Periodontol Res 1977;12:444–449.

[11] Chen M, Xu H. Dysregulation in Retinal Para-Inflammation and Age-Related Retinal

Degeneration in CCL2 or CCR2 Deficient Mice. PLoS ONE 2011;6:e22818.

Page 61: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

51

[12] Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Progress in Retinal and

Eye Research 2009;28:348–368.

[13] Chovatiya R, Medzhitov R. Stress, inflammation, and defense of homeostasis. Molecular

Cell Review 2014;54:281–288.

[14] Medzhitov R. Origin and physiological roles of inflammation. Nature 2008;454:428–435.

[15] Omori K, Ohira T, Uchida Y, Ayilavarapu S, Batista EL, Yagi M, et al. Priming of

neutrophil oxidative burst in diabetes requires preassembly of the NADPH oxidase. J Leukoc

Biol 2008;84:292–301.

[16] Lakschevitz FS, Aboodi GM, Glogauer M. Oral neutrophil transcriptome changes result in a

pro-survival phenotype in periodontal diseases. PLoS ONE 2013;8:e68983.

[17] Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, et al. A distinct

subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus

induces vascular damage and synthesizes type I IFNs. J Immunol 2010;184:3284–3297.

[18] Carmona-Rivera C, Kaplan MJ. Low-density granulocytes: a distinct class of neutrophils in

systemic autoimmunity. Semin Immunopathol 2013;35:455–463.

[19] Sagiv JY, Michaeli J, Assi S, Mishalian I, Kisos H, Levy L, et al. Phenotypic Diversity and

Plasticity in Circulating Neutrophil Subpopulations in Cancer. Cell Reports 2015;10:562–573.

[20] Medzhitov R. Inflammation 2010: New Adventures of an Old Flame. Cell 2010;140:771.

[21] Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat

Immunol 2005;6:1191–1197.

[22] Gold PW, Licinio J, Pavlatou MG. Pathological parainflammation and endoplasmic

reticulum stress in depression: potential translational targets through the CNS insulin, klotho and

PPAR-γ systems. Molecular Psychiatry 2012;18:154.

[23] Gregor MF, Hotamisligil GS, haff. Inflammatory mechanisms in obesity. Annu Rev

Immunol 2011;29:415–445.

Page 62: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

52

[24] Costalonga M. The oral microbiome and the immunobiology of periodontal disease and

caries. Immunology Letters 2014;162:22.

[25] Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human

microbiome project. Nature 2007;449:804–810.

[26] Rescigno M. Gut commensal flora: tolerance and homeostasis. F1000 Biol Rep 2009;1:9.

[27] Garrett WS, Gordon JI, Glimcher LH. Homeostasis and inflammation in the intestine. Cell

2010;140:859–870.

[28] Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of

commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell

2004;118:229–241.

[29] Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells carrying

commensal bacteria. Science 2004;303:1662–1665.

[30] SOCRANSKY SS, HAFFAJEE AD. Periodontal microbial ecology. Periodontol 2000

2005;38:135–187.

[31] Hajishengallis G. Periodontitis: from microbial immune subversion to systemic

inflammation. Nature Publishing Group 2015;15:30–44.

[32] Berezow AB, Darveau RP. Microbial shift and periodontitis. Periodontol 2000 2010;55:36–

47.

[33] Marsh PD. Microbial ecology of dental plaque and its significance in health and disease.

Adv Dent Res 1994;8:263–271.

[34] Cebra JJ. Influences of microbiota on intestinal immune system development. The

American Journal of Clinical Nutrition 1999;69:1046S–1051S.

[35] Roberts FA, Darveau RP. Microbial protection and virulence in periodontal tissue as a

function of polymicrobial communities: symbiosis and dysbiosis. Periodontol 2000 2015;69:18–

27.

Page 63: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

53

[36] Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the

immune system. Nature Reviews Immunology 2004;4:478–485.

[37] Hajishengallis G, Liang S, Payne MA, Hashim A, Jotwani R, Eskan MA, et al. Low-

Abundance Biofilm Species Orchestrates Inflammatory Periodontal Disease through the

Commensal Microbiota and Complement. Cell Host &Amp; Microbe 2011;10:497–506.

[38] Löe H, Anerud A, Boysen H, Morrison E. Natural history of periodontal disease in man.

Rapid, moderate and no loss of attachment in Sri Lankan laborers 14 to 46 years of age. J Clin

Periodontol 1986;13:431–445.

[39] Johnson GK, Hill M. Cigarette smoking and the periodontal patient. J Periodontol

2004;75:196–209.

[40] Mealey BL, Oates TW. Diabetes mellitus and periodontal diseases. J Periodontol

2006;77:1289–1303.

[41] Kornman KS, Crane A, Wang HY, di Giovine FS, Newman MG, Pirk FW, et al. The

interleukin-1 genotype as a severity factor in adult periodontal disease. J Clin Periodontol

1997;24:72–77.

[42] Michalowicz BS, Diehl SR, Gunsolley JC, Sparks BS, Brooks CN, Koertge TE, et al.

Evidence of a substantial genetic basis for risk of adult periodontitis. J Periodontol

2000;71:1699–1707.

[43] Fine N, Hassanpour S, Borenstein A, Sima C, Oveisi M, Scholey J, et al. Distinct Oral

Neutrophil Subsets Define Health and Periodontal Disease States. J Dent Res 2016;95:931–938.

[44] Lakschevitz FS, Hassanpour S, Rubin A, Fine N, Sun C. Identification of neutrophil surface

marker changes in health and inflammation using high-throughput screening flow cytometry.

Exp Cell Res 2016;342:200–209.

[45] Hajishengallis E, Hajishengallis G. Neutrophil homeostasis and periodontal health in

children and adults. J Dent Res 2014;93:231–237.

Page 64: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

54

[46] Kobayashi SD, DeLeo FR. Role of neutrophils in innate immunity: a systems biology-level

approach. Wiley Interdisciplinary Reviews: Systems Biology and Medicine 2010;1:309–333.

[47] Borregaard N. Neutrophils, from marrow to microbes. Immunity 2010;33:657–670.

[48] Bainton DF, Ullyot JL, Farquhar MG. The development of neutrophilic polymorphonuclear

leukocytes in human bone marrow. J Exp Med 1971;134:907–934.

[49] Vietinghoff von S, Ley K. Homeostatic Regulation of Blood Neutrophil Counts. The

Journal of Immunology 2008;181:5183–5188.

[50] Laterveer L, Heemskerk DP, Camps JA, Pauwels EK, Willemze R, Fibbe WE. Rapid

mobilization of haematopoietic progenitor cells in Rhesus monkeys by a single intravenous

injection of interleukin-8. Blood 1996;2:232.

[51] Dale DC, Liles WC, Llewellyn C, Price TH. Effects of granulocyte-macrophage colony-

stimulating factor (GM-CSF) on neutrophil kinetics and function in normal human volunteers.

Am J Hematol 1998;57:7–15.

[52] Theilgaard-Mönch K, Jacobsen LC, Borup R, Rasmussen T, Bjerregaard MD, Nielsen FC,

et al. The transcriptional program of terminal granulocytic differentiation. Blood

2004;105:1785–1796.

[53] Eash KJ, Means JM, White DW, Link DC. CXCR4 is a key regulator of neutrophil release

from the bone marrow under basal and stress granulopoiesis conditions. Blood 2009;113:4711–

4719.

[54] Hernandez PA, Gorlin RJ, Lukens JN, Taniuchi S, Bohinjec J, Francois F, et al. Mutations

in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined

immunodeficiency disease. Nature Genetics 2003;34:70–74.

[55] Borregaard N, Kjeldsen L, Sengeløv H, Diamond MS, Springer TA, Anderson HC, 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

1994;56:80–87.

Page 65: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

55

[56] Eash KJ, Greenbaum AM, Gopalan PK, Link DC. CXCR2 and CXCR4 antagonistically

regulate neutrophil trafficking from murine bone marrow. J Clin Invest 2010;120:2423–2431.

[57] Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic

neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 2005;22:285–294.

[58] Vietinghoff von S, Ley K. IL-17A controls IL-17F production and maintains blood

neutrophil counts in mice. J Immunol 2009;183:865–873.

[59] Mcdonald PP. Transcriptional Regulation in Neutrophils: Teaching Old Cells New Tricks.

vol. 82. 2004.

[60] Cronkite EP, Fliedner TM. GRANULOCYTOPOIESIS. N Engl J Med 1964;270:1403–

1408.

[61] Barletta KE, Cagnina RE, Wallace KL, Ramos SI, Mehrad B, Linden J. Leukocyte

compartments in the mouse lung: Distinguishing between marginated, interstitial, and alveolar

cells in response to injury. J Immunol Methods 2012;375:100–110.

[62] Steele RW, Steele CR, Pilkington NS, Charlton RK. Functional capacity of marginated and

bone marrow reserve granulocytes. Infect Immun 1987;55:2359–2363.

[63] Fliedner TM, Cronkite EP, Killmann SÅ, Bond VP. Granulocytopoiesis. II. Emergence and

pattern of labeling of neutrophilic granulocytes in humans. Blood 1964;24:683–700.

[64] Kim M-H, Granick JL, Kwok C, Walker NJ, Borjesson DL, Curry F-RE, et al. Neutrophil

survival and c-kit(+)-progenitor proliferation in Staphylococcus aureus-infected skin wounds

promote resolution. Blood 2011;117:3343–3352.

[65] Hirai H, Zhang P, Dayaram T, Hetherington CJ, Mizuno S-I, Imanishi J, et al. C/EBPbeta is

required for “emergency” granulopoiesis. Nat Immunol 2006;7:732–739.

[66] Misselwitz B, Bachli EB, Kaiser P, Fehr J, Goede JS. Diagnosis of hypersplenism with the

epinephrine stimulation test - 23 years of experience at a tertiary care hospital. Swiss Med Wkly

2012;141:w13324.

Page 66: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

56

[67] BIERMAN HR, KELLY KH, KING FW, PETRAKIS NL. The Pulmonary Circulation as a

Source of Leucocytes and Platelets in Man. Science 1951;114:276.

[68] Lawrence MB, Springer TA. Leukocytes roll on a selectin at physiologic flow rates:

Distinction from and prerequisite for adhesion through integrins. Cell 1991;65:859.

[69] Ramachandran V, Williams M, Yago T, Schmidtke DW, McEver RP. Dynamic alterations

of membrane tethers stabilize leukocyte rolling on P-selectin. Proc Natl Acad Sci USA

2004;101:13519–13524.

[70] Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the

leukocyte adhesion cascade updated. Nature Reviews Immunology 2007;7:678–689.

[71] Nourshargh S, Hordijk PL, Sixt M. Breaching multiple barriers: leukocyte motility through

venular walls and the interstitium. Nat Rev Mol Cell Biol 2010;11:366–378.

[72] Choi EY, Chavakis E, Czabanka MA, Langer HF, Fraemohs L, Economopoulou M, et al.

Del-1, an endogenous leukocyte-endothelial adhesion inhibitor, limits inflammatory cell

recruitment. Science 2008;322:1101–1104.

[73] Sundd P, Gutierrez E, Koltsova EK, Kuwano Y, Fukuda S, Pospieszalska MK, et al.

“Slings” enable neutrophil rolling at high shear. Nature 2012;488:399–403.

[74] Woodfin A, Voisin M-B, Nourshargh S. Recent developments and complexities in

neutrophil transmigration. Current Opinion in Hematology 2009;17:9–17.

[75] Cooper D, Lindberg FP, Gamble JR, Brown EJ, Vadas MA. Transendothelial migration of

neutrophils involves integrin-associated protein (CD47). Proceedings of the National Academy

of Sciences 1995;92:3978.

[76] Muller WA. PECAM-1 is required for transendothelial migration of leukocytes. Journal of

Experimental Medicine 1993;178:449.

[77] Muller WA. Mechanisms of Leukocyte Transendothelial Migration. Annu Rev Pathol Mech

Dis 2011;6:323–344.

Page 67: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

57

[78] Diamond MS. ICAM-1 (CD54): a counter-receptor for Mac-1 (CD11b/CD18). The Journal

of Cell Biology 1990;111:3129.

[79] Khan AI, Kerfoot SM, Heit B, Liu L, Andonegui G, Ruffell B, et al. Role of CD44 and

Hyaluronan in Neutrophil Recruitment. The Journal of Immunology 2004;173:7594.

[80] Hyun Y-M, Sumagin R, Sarangi PP, Lomakina E, Overstreet MG, Baker CM, et al. Uropod

elongation is a common final step in leukocyte extravasation through inflamed vessels. J Exp

Med 2012;209:1349–1362.

[81] Briheim G, Coble B, Stendahl O, Dahlgren C. Exudate polymorphonuclear leukocytes

isolated from skin chambers are primed for enhanced response to subsequent stimulation with

chemoattractant f-Met-Leu-Phe and C3-opsonized yeast particles. Inflammation 1988;12:141–

152.

[82] Theilgaard-Mönch K, Knudsen S, Follin P, Borregaard N. The transcriptional activation

program of human neutrophils in skin lesions supports their important role in wound healing.

The Journal of Immunology 2004;172:7684–7693.

[83] Casanova-Acebes M, Pitaval C, Weiss LA, Nombela-Arrieta C, Chèvre R, A-González N, et

al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell

2013;153:1025–1035.

[84] Gamonal J, Sanz M, O'Connor A, Acevedo A, Suarez I, Sanz A, et al. Delayed neutrophil

apoptosis in chronic periodontitis patients. J Clin Periodontol 2003;30:616–623.

[85] Witko-Sarsat V, Rieu P, Descamps-Latscha B, Lesavre P, Halbwachs-Mecarelli L.

Neutrophils: molecules, functions and pathophysiological aspects. Lab Invest 2000;80:617–653.

[86] Buckley CD, Ross EA, McGettrick HM, Osborne CE, Haworth O, Schmutz C, et al.

Identification of a phenotypically and functionally distinct population of long-lived neutrophils

in a model of reverse endothelial migration. J Leukoc Biol 2006;79:303–311.

[87] Haslett C, Savill JS, Whyte MK, Stern M, Dransfield I, Meagher LC. Granulocyte apoptosis

and the control of inflammation. Philos Trans R Soc Lond, B, Biol Sci 1994;345:327–333.

Page 68: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

58

[88] Kobayashi SD, Voyich JM, Whitney AR, DeLeo FR. Spontaneous neutrophil apoptosis and

regulation of cell survival by granulocyte macrophage-colony stimulating factor. J Leukoc Biol

2005;78:1408–1418.

[89] Borregaard N, Sørensen OE, Theilgaard-Mönch K. Neutrophil granules: a library of innate

immunity proteins. Trends Immunol 2007;28:340–345.

[90] Sengeløv H, Kjeldsen L, Borregaard N. Control of exocytosis in early neutrophil activation.

J Immunol 1993;150:1535–1543.

[91] Borregaard N. Granules of the human neutrophilic polymorphonuclear leukocyte. Blood

1997;89:3503–3521.

[92] Simon SI, Cherapanov V, Nadra I, Waddell TK, Seo SM, Wang Q, et al. Signaling functions

of L-selectin in neutrophils: alterations in the cytoskeleton and colocalization with CD18. J

Immunol 1999;163:2891–2901.

[93] Le Cabec V, Cowland JB, 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. Proceedings of the National Academy of

Sciences 1996;93:6454.

[94] Brumell JH, Volchuk A, Sengeløv H, Borregaard N, Cieutat AM, Bainton DF, et al.

Subcellular distribution of docking/fusion proteins in neutrophils, secretory cells with multiple

exocytic compartments. J Immunol 1995;155:5750–5759.

[95] Amulic B, Cazalet C, Hayes GL, Metzler KD, Zychlinsky A. Neutrophil function: from

mechanisms to disease. Ann Rev Immunol 2012;30:459–489.

[96] Dewitt S, Laffafian I, Hallett MB. Phagosomal oxidative activity during beta2 integrin

(CR3)-mediated phagocytosis by neutrophils is triggered by a non-restricted Ca2+ signal: Ca2+

controls time not space. J Cell Sci 2003;116:2857–2865.

[97] Babior BM, Kipnes RS, Curnutte JT. Biological defense mechanisms. The production by

leukocytes of superoxide, a potential bactericidal agent. J Clin Invest 1973;52:741–744.

Page 69: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

59

[98] Quinn MT, Gauss KA. Structure and regulation of the neutrophil respiratory burst oxidase:

comparison with nonphagocyte oxidases. J Leukoc Biol 2004;76:760–781.

[99] Nauseef WM. Biological roles for the NOX family NADPH oxidases. Journal of Biological

Chemistry 2008;283:16961–16965.

[100] Heyworth PG, Cross AR, Curnutte JT. Chronic granulomatous disease. Current Opinion in

Immunology 2003;15:578–584.

[101] Robinson JM, Ohira T, Badwey JA. Regulation of the NADPH-oxidase complex of

phagocytic leukocytes. Recent insights from structural biology, molecular genetics, and

microscopy. Histochemistry and Cell Biology 2004;122:293–304.

[102] Chapple ILC, Matthews JB. The role of reactive oxygen and antioxidant species in

periodontal tissue destruction. Periodontol 2000 2007;43:160–232.

[103] McCormick A, Heesemann L, Wagener J, Marcos V, Hartl D, Loeffler J, et al. NETs

formed by human neutrophils inhibit growth of the pathogenic mold Aspergillus fumigatus.

Microbes and Infection 2010;12:928.

[104] Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs.

Nature Reviews Microbiology 2007;5:577–582.

[105] Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al.

Neutrophil extracellular traps kill bacteria. Science 2004;303:1532–1535.

[106] Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A. Neutrophil elastase and

myeloperoxidase regulate the formation of neutrophil extracellular traps. The Journal of Cell

Biology 2010;191:677–691.

[107] Bianchi M, Hakkim A, Brinkmann V, Siler U, Seger RA, Zychlinsky A, et al. Restoration

of NET formation by gene therapy in CGD controls aspergillosis. Blood 2009;114:2619–2622.

[108] Klebanoff SJ, Kettle AJ, Rosen H, Winterbourn CC, Nauseef WM. Myeloperoxidase: a

front-line defender against phagocytosed microorganisms. J Leukoc Biol 2012;93:185–198.

Page 70: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

60

[109] Haneke E. The Papillon-Lefèvre syndrome: keratosis palmoplantaris with periodontopathy.

Report of a case and review of the cases in the literature. Hum Genet 1979;51:1–35.

[110] Knight JS, Kaplan MJ. Lupus neutrophils: “NET” gain in understanding lupus

pathogenesis. Curr Opin Rheumatol 2012;24:441–450.

[111] Pillay J, Braber den I, Vrisekoop N, Kwast LM, de Boer RJ, Borghans JAM, et al. In vivo

labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood 2010;116:625–627.

[112] Tak T, Tesselaar K, Pillay J, Borghans JAM, Koenderman L. What's your age again?

Determination of human neutrophil half-lives revisited. J Leukoc Biol 2013;94:595–601.

[113] Whyte MK, Meagher LC, MacDermot J, Haslett C. Impairment of function in aging

neutrophils is associated with apoptosis. J Immunol 1993;150:5124–5134.

[114] Beyrau M, Bodkin JV, Nourshargh S. Neutrophil heterogeneity in health and disease: a

revitalized avenue in inflammation and immunity. Open Biol 2012;2:120134–120134.

[115] Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and

regulation of innate and adaptive immunity. Nature Reviews Immunology 2011;11:519–531.

[116] Mócsai A. Diverse novel functions of neutrophils in immunity, inflammation, and beyond.

J Exp Med 2013;210:1283–1299.

[117] Nathan C. Neutrophils and immunity: challenges and opportunities. Nature Reviews

Immunology 2006;6:173–182.

[118] Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis

2012;33:949–955.

[119] Tsuda Y, Takahashi H, Kobayashi M, Hanafusa T, Herndon DN, Suzuki F. Three different

neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-

resistant Staphylococcus aureus. Immunity Review 2004;21:215–226.

[120] Hotchkiss RS, Coopersmith CM, McDunn JE, Ferguson TA. The sepsis seesaw: tilting

toward immunosuppression. Nat Med 2009;15:496–497.

Page 71: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

61

[121] Pillay J, Kamp VM, van Hoffen E, Visser T, Tak T, Lammers J-W, et al. A subset of

neutrophils in human systemic inflammation inhibits T cell responses through Mac-1. J Clin

Invest 2012;122:327–336.

[122] Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, et al. Neutrophils

activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus

erythematosus. Sci Transl Med 2011;3:73ra19.

[123] Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, et al. Crosstalk between

neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med

2012;19:65–73.

[124] Maecker HT, Lindstrom TM, Robinson WH, Utz PJ, Hale M, Boyd SD, et al. New tools

for classification and monitoring of autoimmune diseases. Nature Reviews Rheumatology

2012;8:317–328.

[125] Dumitru CA, Moses K, Trellakis S, Lang S. Neutrophils and granulocytic myeloid-derived

suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology.

Cancer Immunology 2012;61:1155–1167.

[126] Fung E, Esposito L, Todd JA, Wicker LS. Multiplexed immunophenotyping of human

antigen-presenting cells in whole blood by polychromatic flow cytometry. Nat Protoc

2010;5:357–370.

[127] Brooks CR, van Dalen CJ, Hermans IF, Douwes J. Identifying leukocyte populations in

fresh and cryopreserved sputum using flow cytometry. Cytometry Part B: Clinical Cytometry

2013;84:104–113.

[128] Fortunati E, Kazemier KM, Grutters JC, Koenderman L, Van den Bosch VJMM. Human

neutrophils switch to an activated phenotype after homing to the lung irrespective of

inflammatory disease. Clin Exp Immunol 2008;155:559–566.

[129] Tak T, Hilvering B, Tesselaar K, Koenderman L. Similar activation state of neutrophils in

sputum of asthma patients irrespective of sputum eosinophilia. Clin Exp Immunol

2015;182:204–212.

Page 72: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

62

[130] Quinn MT, Deleo F, Bokoch GM. Neutrophil methods and protocols. 2007.

[131] Kuijpers TW, Tool AT, van der Schoot CE, Ginsel LA, Onderwater JJ, Roos D, et al.

Membrane surface antigen expression on neutrophils: a reappraisal of the use of surface markers

for neutrophil activation. Blood 1991;78:1105–1111.

[132] Colgan SP, Ehrentraut SF, Glover LE, Kominsky DJ, Campbell EL. Contributions of

neutrophils to resolution of mucosal inflammation. Immunol Res 2012;55:75–82.

[133] Fournier BM, Parkos CA. The role of neutrophils during intestinal inflammation. Mucosal

Immunology 2012;5:354–366.

[134] Kühl AA, Kakirman H, Janotta M, Dreher S, Cremer P, Pawlowski NN, et al. Aggravation

of different types of experimental colitis by depletion or adhesion blockade of neutrophils.

Gastroenterology 2007;133:1882–1892.

[135] Calouius PEB. The leukocyte count in saliva. Oral Surgery, Oral Medicine, Oral Pathology

1958;11:43.

[136] Raeste AM. Degeneration of oral leukocytes. Eur J Oral Sci 1972;80:285–291.

[137] Nussbaum G, Shapira L. How has neutrophil research improved our understanding of

periodontal pathogenesis? J Clin Periodontol 2011;38 Suppl 11:49–59.

[138] Landzberg M, Doering H, Aboodi GM, Tenenbaum HC, Glogauer M. Quantifying oral

inflammatory load: oral neutrophil counts in periodontal health and disease. J Periodontal Res

2014;50:330–336.

[139] Davison G, Jones AW. Oral neutrophil responses to acute prolonged exercise may not be

representative of blood neutrophil responses. Appl Physiol Nutr Metab 2014;40:1–4.

[140] Rovin S. The influence of bacteria and irritation in the initiation of periodontal disease in

germfree and conventional rats. J Periodontal Res 1966;1:193.

Page 73: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

63

[141] Tonetti MS, Imboden MA, Lang NP. Neutrophil migration into the gingival sulcus is

associated with transepithelial gradients of interleukin-8 and ICAM-1. J Periodontol

1998;69:1139–1147.

[142] Page RC, Schroeder HE. Current Status of the Host Response in Chronic Marginal

Periodontitis*. J Periodontol 1981;52:477.

[143] Delima AJ, Van Dyke TE. Origin and function of the cellular components in gingival

crevice fluid. Periodontol 2000 2003;31:55–76.

[144] Schiott CR, Loe H. The origin and variation in number of leukocytes in the human saliva. J

Periodontal Res 1970;5:36–41.

[145] Deas DE, Mackey SA, McDonnell HT. Systemic disease and periodontitis: manifestations

of neutrophil dysfunction. Periodontol 2000 2003;32:82.

[146] Fredriksson M, Gustafsson A, Asman B, Bergstrom K. Hyper-reactive peripheral

neutrophils in adult periodontitis: generation of chemiluminescence and intracellular hydrogen

peroxide after in vitro priming and FcgammaR-stimulation. J Clin Periodontol 1998;25:394.

[147] Lakschevitz FS, Aboodi GM, Glogauer M. Oral neutrophils display a site-specific

phenotype characterized by expression of T-cell receptors. J Periodontol 2013;84:1493–1503.

[148] Guarnieri C, Zucchelli G, Bernardi F, Scheda M, Valentini AF, Calandriello M. Enhanced

Superoxide Production with on Change of the Antioxidant Activity in Gingival Fluid of Patients

with Chronic Adult Periodontitis. Free Radical Research Communications 1991;15:11.

[149] Niture SK, Khatri R, Jaiswal AK. Regulation of Nrf2-an update. Free Radic Biol Med

2013;66:36–44.

[150] Armitage GC. Development of a classification system for periodontal diseases and

conditions. Ann Periodontol 1999;4:1–6.

[151] Lang NP, Adler R, Joss A, Nyman S. Absence of bleeding on probing. An indicator of

periodontal stability. J Clin Periodontol 1990;17:714–721.

Page 74: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

64

[152] Mancini S, Romanelli R, Laschinger CA, Overall CM, Sodek J, McCulloch CAG.

Assessment of a Novel Screening Test for Neutrophil Collagenase Activity in the Diagnosis of

Periodontal Diseases. J Periodontol 1999;70:1292–1302.

[153] Ashkenazi M, Dennison DK. A New Method for Isolation of Salivary Neutrophils and

Determination of Their Functional Activity. J Dent Res 1989;68:1256–1261.

[154] Lim MBH, Kuiper JWP, Katchky A, Goldberg H, Glogauer M. Rac2 is required for the

formation of neutrophil extracellular traps. J Leukoc Biol 2011;90:771–776.

[155] Coulter HD. Rapid and improved methods for embedding biological tissues in Epon 812

and Araldite 502. Journal of Ultrastructure Research 1967;20:346–355.

[156] Gavillet M, Martinod K, Renella R, Harris C, Shapiro NI, Wagner DD, et al. Flow

cytometric assay for direct quantification of neutrophil extracellular traps in blood samples. Am J

Hematol 2015;90:1155–1158.

[157] Passlick B, Flieger D, Ziegler-Heitbrock HW. Identification and characterization of a novel

monocyte subpopulation in human peripheral blood. Blood 1989;74:2527–2534.

[158] Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild T, Dutzan N, et al. Defective

neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven

inflammatory bone loss. Sci Transl Med 2014;6:229ra40.

[159] Kantarci A, Oyaizu K, Van Dyke DTE. Neutrophil-Mediated Tissue Injury in Periodontal

Disease Pathogenesis: Findings from Localized Aggressive Periodontitis. J Periodontol

2003;74:66–75.

[160] Jimenez-Flores E. Human neutrophils and oral microbiota: a constant tug-of-war between a

harmonious and a discordant coexistence. Immunol Rev 2016;273:282.

[161] Cloke T, Munder M, Taylor G, Müller I, Kropf P. Characterization of a novel population

of low-density granulocytes associated with disease severity in HIV-1 infection. PLoS ONE

2012;7:e48939.

Page 75: Distinct Oral Neutrophil Subsets Define Health and Disease ... · Siavash Hassanpour Master of Science Faculty of Dentistry University of Toronto 2017 Abstract Neutrophils exit the

65

[162] Elghetany MT. Surface Antigen Changes during Normal Neutrophilic Development: A

Critical Review. Blood Cells, Molecules, and Diseases 2002;28:260.

[163] van Eeden SF, Klut ME, Walker BAM, Hogg JC. The use of flow cytometry to measure

neutrophil function. J Immunol Methods 1999;232:23–43.

[164] Kobuch J, Cui H, Grünwald B, Saftig P, Knolle PA, Kruger A. TIMP-1 signaling via

CD63 triggers granulopoiesis and neutrophilia in mice . Hematopoiesis 2015;100:1005–1013.

[165] Cham BP, Gerrard JM, Bainton DF. Granulophysin is located in the membrane of

azurophilic granules in human neutrophils and mobilizes to the plasma membrane following cell

stimulation. Am J Pathol 1994;144:1369–1380.

[166] Streimish I, Bizzarro M, Northrup V, Wang C, Renna S, N K, et al. Neutrophil CD64 as a

diagnostic marker of sepsis in neonates. The Pediatric Infecious Disease Journal 2012;31:777–

781.

[167] Hoffmann J. Neutrophil CD64: a diagnostic marker for infection and sepsis. Clinical

Chemistry and Laboratory Medicine 2009;47:903–916.

[168] Skubitz KM, Campbell KD, Skubitz AP. CD66a, CD66b, CD66c, and CD66d each

independently stimulate neutrophils. J Leukoc Biol 1996;60:106–117.

[169] Christmas SE, La Mata Espinosa De CT, Halliday D, Buxton CA, Cummerson JA, Johson

PM. Levels of expression of complement regulatory proteins CD46, CD55 and CD59 on resting

and activated human peripheral blood leucocytes. Immunology 2006;119:522–528.

[170] Avril T, Freeman SD, Attrill H, Clarke RG, Crocker PR. Siglec-5 (CD170) can mediate

inhibitory signaling in the absence of immunoreceptor tyrosine-based inhibitory motif

phosphorylation. J Biol Chem 2005;280:19843–19851.