107
The Effect of Helicobacter pylori on Innate Immunity by Michelle Ang A thesis submitted in conformity with the requirements for the degree of Masters of Science Graduate Department of Physiology University of Toronto © Copyright by Michelle Ang (2010)

Ang Michelle 201006 MSc Thesis

Embed Size (px)

Citation preview

Page 1: Ang Michelle 201006 MSc Thesis

The Effect of Helicobacter pylori on Innate Immunity

by

Michelle Ang

A thesis submitted in conformity with the requirements

for the degree of Masters of Science

Graduate Department of Physiology

University of Toronto

© Copyright by Michelle Ang (2010)

Page 2: Ang Michelle 201006 MSc Thesis

ii

The Effect of Helicobacter pylori on Innate Immunity

Michelle Ang

Master of Science

Department of Physiology

University of Toronto

2010

Abstract

The innate immune system is important in both acute and chronic infection. In this thesis, I

investigated the effect of H. pylori infection on 1) DCs, key orchestrators of the immune system,

and 2) autophagy, recently identified as an important component of innate immunity. I

determined that H. pylori activates the STAT3 pathway in DCs, increasing DC maturation and

inducing production of IL-10, IL-12p40 and TNF-α, without IL-12p70. This cytokine profile

may favour an immunoregulatory response, promoting persistent H. pylori infection. In addition

I determined that H. pylori’s VacA toxin induced autophagy, ROS production and Parkin

aggregation which has been implicated in mediating autophagy in response to mitochondrial

damage. Thus H. pylori alters these key effectors of innate immunity which may play a role in

promoting its chronic infection and disease.

Page 3: Ang Michelle 201006 MSc Thesis

iii

Acknowledgments

The many people who have helped me through the graduate process are so numerous that to

name them all would double this thesis’s length. However, I would like to acknowledge several

key people to whom I am greatly indebted.

To my lab members, thank you for your unwavering support, and for the amazing environment in

which I was privileged enough to work. Thank you Esther. Your sense of fun and humour were

integral to the great atmosphere that exists in the lab. Thank you for helping with me with all my

mice, for all the great advice, and all the amazing talks about food. To Deepa, thank you for all

the help with the autophagy project. I will miss our daily chats about cricket, world news and

sports, and I thank you for letting me rant when experiments experienced catastrophic failure!

Dana, for making the lab fun, for teaching me most of the basics when I first started, and for

allowing me to work on your great paper.

To my supervisor, Dr. Nicola Jones, for the tremendous mentorship, helping me reach levels in

research I could not have done on my own. Thank you for caring not only about the results, but

about making science enjoyable for me as a person. It is rare to find a supervisor who cares not

only about the data, but about the well being and stress levels of the person and I thank you for

helping me through the difficult times in my Masters career.

Thank you to my committee members, Dr. Dana Philpott and Dr. Michael Wheeler. You have

both given me great feedback and guidance, and I thank you for your mentorship throughout this

process.

I’d like to thank the Philpott lab, specifically Joao and Thiru, who listened to millions of

questions and provided countless hours of support in my FACS data.

To rock climbing! For being the best outlet for experimental aggression I could have ever hoped

for.

To my family, thank you for being there for me when I needed you. To my Ahmah, who will

never read a word of this language, but whose unconditional love and support I cherish with all

my heart.

Page 4: Ang Michelle 201006 MSc Thesis

iv

To my wonderful friends. You all know who you are. I love you all dearly, and am inspired by

all your successes. I am lucky to have such lovely and intelligent people in my life.

To my Greg, without whom I would never have completed this stage in my life. You have been

my number one supporter, having more confidence in me than I ever could have in myself. I

cannot express how much your love and support has meant to me. Thank you for holding my

hand through the tough times in the lab, for encouraging me, and for saying all the things I

needed to hear to find the will to finish. I could not have done this without you. Thank you, and I

love you.

And to God, for knowing I can turn to you when I need to.

Page 5: Ang Michelle 201006 MSc Thesis

v

Table of Contents

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

Table of Contents ............................................................................................................................ v

List of Abbreviations .................................................................................................................... vii

List of Tables ................................................................................................................................. xi

List of Figures ............................................................................................................................... xii

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

1 Helicobacter pylori .................................................................................................................... 1

1.1 Infection and Colonization .................................................................................................. 1

1.2 H. pylori Associated Diseases ............................................................................................. 2

1.3 H. pylori Virulence Factors ................................................................................................. 2

1.3.1 Cytotoxin Associated Gene A (CagA) .................................................................... 2

1.3.2 Vacuolating Cytotoxin A (VacA) ........................................................................... 4

1.3.3 Lipopolysaccharide (LPS) .................................................................................... 13

2 Innate Immunity ....................................................................................................................... 13

2.1 Dendritic Cells (DCs) ....................................................................................................... 14

2.2 Signal Transducer and Activator of Transcription 3 (STAT3) ......................................... 18

2.2.1 STAT3 Signalling Cascade ................................................................................... 18

2.2.2 STAT3, DCs and Cancer ...................................................................................... 21

2.2.3 STAT3 and H. pylori ............................................................................................ 21

2.3 Dendritic Cells and H. pylori ............................................................................................ 21

2.4 Autophagy ......................................................................................................................... 26

2.4.1 H. pylori and Autophagy ....................................................................................... 31

3 Summary .................................................................................................................................. 31

Chapter 2 Materials and Methods ................................................................................................. 33

Page 6: Ang Michelle 201006 MSc Thesis

vi

CHAPTER 3 MATERIALS AND METHODS ....................................................................... 33

CHAPTER 4 MATERIALS AND METHODS ....................................................................... 35

Chapter 3 H. pylori Activates the STAT3 Pathway in DCs, Causing DC Maturation but

Altering DC Cytokine Release ................................................................................................. 39

ABSTRACT ............................................................................................................................. 39

INTRODUCTION.................................................................................................................... 39

RESULTS ................................................................................................................................ 41

H. pylori infection induces tyrosine phosphorylation of STAT3 ..................................... 41

H. pylori induces expression of DC maturation markers .................................................. 44

H. pylori stimulated DCs induce an unusual cytokine profile .......................................... 47

H. pylori induced DC maturation and cytokine release is dependent on activation of

the STAT3 pathway. ............................................................................................. 52

S3I-201 does not affect DC viability. ............................................................................... 55

DISCUSSION .......................................................................................................................... 58

Chapter 4 Helicobacter pylori’s VacA Toxin Induces Reactive Oxygen Species Production

and Alters Parkin Distribution ................................................................................................. 61

INTRODUCTION.................................................................................................................... 61

RESULTS ................................................................................................................................ 62

H. pylori’s VacA toxin induces ROS production ............................................................. 62

VacA induced autophagosomes do not colocalize with the mitochondria ....................... 68

H. pylori’s VacA toxin causes Parkin redistribution that is not dependent on VacA

induced autophagy ................................................................................................ 71

VacA induced Parkin aggregation does not colocalize with the mitochondria. ............... 74

DISCUSSION .......................................................................................................................... 77

Chapter 5 Discussion and Future Directions ................................................................................ 79

FUTURE DIRECTIONS ......................................................................................................... 81

References ..................................................................................................................................... 83

Page 7: Ang Michelle 201006 MSc Thesis

vii

List of Abbreviations

APC Antigen presenting cell

Atg Autophagy gene

BMDC Bone Marrow derived Dendritic Cells

CagA Cytotoxin Associated Gene A

cagPAI cag Pathogenicity Island

CD Cluster of Differentiation

CKO Conditional knock-out

DC Dendritic Cell

DNA Deoxyribonucleic acid

DPI diphenylene iodonium

ELISA Enzyme-Linked Immunosorbent Assay

EPIYA Glu-Pro-Ile-Tyr-Ala

FACS Fluorescence Activated Cell Sorting

FBS Fetal Bovine Serum

FITC Flourescein isothiocyanate

GAS Gamma-IFN–stimulated activation sequence

GEEC GPI-AP-enriched early endosomal compartments

GFP Green Fluorescent Protein

GM-CSF Granulocyte-macrophage colony-stimulating factor

Page 8: Ang Michelle 201006 MSc Thesis

viii

HK Heat Killed WT bacteria

H. pylori Helicobacter pylori

IBD Inflammatory Bowel Disease

IFN Interferon

IL Interleukin

ISRE IFNα-stimulated gene response element

JAK Janus Kinase

JAM Junctional adhesion molecule

LBP LPS binding protein

LC3 Mictotubule-associated Light Chain 3

LPS Lipopolysaccharide

MAP Mitogen-Activated Protein

MHC Major Histocompatibility Complex

Min Minute

MOI Multiplicities of Infection

MPT Mitochondrial Permeability Transition

mTOR Mammalian Target of Rapamycin

MyD88 Myeloid Differentiation primary response gene 88

NAC N-acetylcysteine

NADPH Nicotinamide adenine dinucleotide phosphate

Page 9: Ang Michelle 201006 MSc Thesis

ix

NAP Neutrophil Activating Protein

NBT Nitroblue tetrazolium

NO Nitric Oxide

NF-κB Nuclear factor kappa-light-chain enhancer of activated B-cells

PAS Pre-Autophagosomal Structures

PAMP Pathogen-Associated Molecular Patterns

PE Posphatidylethanolamine

PerCP-Cy5.5 Peridinin chlorophyll protein conjugated to cyanine

PI3K Phosphatidylinositol-3-kinases

PSTAT3 Phosphorylated STAT3

RFP Red Fluorescent Protein

RNA Ribonucleic acid

ROS Reactive Oxygen Species

RPMI Roswell Park Memorial Institute

SH2 Src-Homology 2

siRNA Small interfering RNA

SNARE Soluble NSF Attachment Receptor

SNAP Soluble N-ethylmaleimide-sensitive attachment protein

STAT Signal Transducer and Activator of Transcription

TBST Tris-buffered saline with tween

Page 10: Ang Michelle 201006 MSc Thesis

x

Th T-helper

TFSS Type IV Secretion System

TGF Tumour Growth Factor

TLR Toll-Like Receptor

TNF-α Tumour Necrosis Factor-alpha

Treg T-regulatory cells

VacA Vacoulating Cytotoxin A

WT Wild-type

YFP Yellow Fluorescent protein

Page 11: Ang Michelle 201006 MSc Thesis

xi

List of Tables

Table 1: Host, genetic bacterial and environmental factors in the pathogenesis of H. pylori

induced gastric cancer.

Page 12: Ang Michelle 201006 MSc Thesis

xii

List of Figures

Figure 1-1: H. pylori CagA signalling pathway

Figure 1-2: VacA induces cellular vacuolation.

Figure 1-3: Multiple actions of H. pylori’s VacA Toxin

Figure 1-4: Model of DC maturation

Figure 1-5: The STAT3 activation pathway

Figure 1-6: From Bronte-Tinkew et al. Nuclear STAT3 detected in epithelial and inflammatory

cells of H. pylori CagA+ infected Mongolian gerbil tissue

Figure 1-7: Summary of the possible immune responses to H. pylori

Figure 1-8: Overview of the autophagic process in mammalian cells

Figure 3-1: H. pylori activates the STAT3 activation pathway in DCs

Figure 3-2: H. pylori infection upregulates surface expression of DC maturation markers

Figure 3-3: Luminex assay of H. pylori induced by DC secretion

Figure 3-4: ELISA determination of H. pylori induced DC cytokine secretion

Figure 3-5: H.pylori induced DC maturation and cytokine secretion is dependent on activation of

the STAT3 pathway

Figure 3-6: S3I-201 does not affect BMDC viability

Figure 4-1: VacA pure toxin induces ROS production in AGS cells.

Figure 4-2: VacA induced ROS production is not responsible for VacA induced autophagy

Figure 4-3: VacA induced autophagy does not colocalize with mitochondrial antibodies

Page 13: Ang Michelle 201006 MSc Thesis

xiii

Figure 4-4: VacA pure toxin induces redistribution of Parkin that is independent of its induction

of autophagy

Figure 4-5: VacA induced Parkin aggregation does not target to the mitochondria

Page 14: Ang Michelle 201006 MSc Thesis

1

Chapter 1 Introduction

1 Helicobacter pylori

1.1 Infection and Colonization

Helicobacter pylori (H. pylori) is a gram-negative, spiral shaped bacterium that infects the

stomach of approximately half of the world’s human population(Kusters et al., 2006). H. pylori

is acquired early in life and persists for the lifetime of the host. Infection is associated with the

development of chronic gastritis and peptic ulcers (Cover and Blaser, 2009). In addition, H.

pylori is classified as a type 1 carcinogen and is the single most important risk factor in the

development of gastric cancer, the second most common cancer worldwide (Amieva and El

Omar, 2008;Correa and Houghton, 2007).

The bacterium is highly adapted to colonize in the human stomach where various factors

including gastric acidity, nutrient availability, innate and adaptive immunity limit most other

bacterial colonization (Cover and Blaser, 2009). H. pylori achieves gastric colonization through a

number of factors such as utilizing flagella which act as propellers to travel through the viscous

mucosa, and through the enzyme urease, which converts urea into NH3 and CO2, thereby

increasing the pH to buffer the environment surrounding the bacteria (Dhar et al.,

2003;Montecucco and Rappuoli, 2001;Portal-Celhay and Perez-Perez, 2006). H. pylori also

utilizes specialized adherence mechanisms ensuring a persistent and chronic infection. Utilizing

the outer-membrane protein BabA, H. pylori strongly adheres to the Lewis B blood group

antigens expressed on gastric epithelial cells and within the gastric mucus (Linden et al.,

2002;Montecucco and Rappuoli, 2001). Another bacterial adhesion SabA also mediates

interaction with Lewis X receptors, maintaining a weaker interaction that is upregulated during

inflammation (Linden et al., 2002). Using these factors, H. pylori adheres to gastric epithelial

cells allowing the bacterium to maintain close proximity to the epithelial cell layer, allowing for

the translocation of bacterial effector proteins and access to cellular nutrients thereby forming an

environmentally favourable niche for bacterial survival.

Page 15: Ang Michelle 201006 MSc Thesis

2

1.2 H. pylori Associated Diseases

Infection with H. pylori is associated with the development of several disease phenotypes. All

individuals infected with H. pylori develop chronic gastritis, an inflammatory response that is

insufficient to eradicate the organism from the host. Additionally, approximately 10-20% of

infected individuals will go on to develop peptic ulcers, associated with antral predominant

gastritis and increased acid secretion (Kusters et al., 2006).

H. pylori infection is also the strongest risk factor for the development of gastric cancers

including lymphoma, MALT lymphoma and adenocarcinoma. Gastric cancer occurs in 1-2% of

the infected population (Cover and Blaser, 2009;Lochhead and El Omar, 2007). Correa proposed

a model of H. pylori-mediated gastric carcinogenesis in which there is a sequential array of

pathological changes that convert normal gastric mucosa to a cancerous environment beginning

with the development of chronic active gastritis, followed by atrophy of the gastric tissue and

inflammatory damage. This causes progression to intestinal metaplasia, dysplasia and eventually

the development of gastric cancer (Correa, 2003;Correa, 2004;Correa and Houghton, 2007).

However, the exact mechanisms leading to the development of gastric cancer remain unclear.

Bacterial, host and environmental factors (Table 1) are thought to contribute to an increased risk

for the development of gastric cancer. Investigating the role of bacterial factors in altering host

immune responses is the focus of my research.

1.3 H. pylori Virulence Factors

1.3.1 Cytotoxin Associated Gene A (CagA)

The most well characterized virulence factor is the cytotoxin-associated gene A (CagA) found

within the (cag) pathogenicity island (PAI). The cag PAI encodes a type IV secretion system

(TFSS) similar to those found in Agrobacterium tumefaciens, Bordetella pertussis and

Legionella pneumophila. The TFSS acts as a molecular syringe to inject CagA proteins into host

cells (Bourzac and Guillemin, 2005;Odenbreit et al., 2000). Infection with strains of H. pylori

containing CagA confers a higher risk for the development of gastric cancer (Parsonnet et al.,

1997). Furthermore transgenic mice expressing the CagA virulence factor develop

gastrointestinal and hematopoietic neoplasms, confirming that CagA is tumourigenic (Ohnishi et

al., 2008). The exact mechanisms by which CagA promotes cancer remain unknown. Once

Page 16: Ang Michelle 201006 MSc Thesis

3

Table 1: Host, genetic bacterial and environmental factors in the pathogenesis of H. pylori

induced gastric cancer.

Host genetic factors Bacterial virulence

factors

Environmental

factors

Gastric cancer

phenotype

IL-1B-511*T

IL-1-RN*2*2

IL-10 ATA haplotype

IL-8-251*A

TLR4+896*G

MBL2 HYD

haplotype

cagPAI, CagA

VacA

Smoking

Poor Diet

Corpus-predominant

gastritis

Multifocal atrophic

gastritis

High gastrin +

Hypochlorhydria

Low pepsinogen I and

pepsinogenI/II ratio

Increased risk of

gastric cancer

Modified from (Amieva and El Omar, 2008).

Page 17: Ang Michelle 201006 MSc Thesis

4

CagA is translocated into host cells via the TFSS, it localizes to the plasma membrane and

undergoes phosphorylation by the SRC family of kinases (Hatakeyama, 2004) (Figure 1-1). This

phosphorylation occurs within a repeated specific 5 amino acid motif (EPIYA – Glu-Pro-Ile-Tyr-

Ala) on the tyrosine residue (Parsonnet et al., 1997). The presence and number or EPIYA repeats

may contribute to severity of disease outcome (Dhar et al., 2003). Upon phosphorylation, CagA

binds to SRC Homology 2 (SH2) domain containing protein tyrosine phosphatase SHP2,

deregulating this oncoprotein, activating phosphatase activity and forming a complex that

initiates mitogenic signalling, cell scattering, elongation and rearrangement of the actin

cytoskeleton (Kusters et al., 2006). CagA also has actions independent of its EPIYA tyrosine

phosphorylation. For example, unphosphorylated CagA can target the c-Met receptor as well as

bind to Grb2 (Churin et al., 2003;Mimuro et al., 2002). CagA also interacts in a phosphorylation

independent manner with ZO-1 and junctional adhesion molecule (JAM) thereby altering tight

junctions (Amieva et al., 2003;Hatakeyama, 2006).

1.3.2 Vacuolating Cytotoxin A (VacA)

Another important bacterial virulence factor is the vacuolating cytotoxin A (VacA) so named for

its ability to induce vacuoles in cells. VacA is a toxin secreted by H. pylori. VacA released from

H. pylori is cleaved into two fragments, a 33 kDa N-terminal fragment (p33) and a 55 kDa c-

terminal fragment which exist as a large ogliomeric complex when purified (Dhar et al., 2003).

Exposure to acidic and alkaline conditions causes these oligomeric components to disassociate

into the monomeric subunits (Cover and Blanke, 2005). The p55 fragment has an important role

in the binding of VacA to host cells (Cover and Blanke, 2005;Galmiche et al., 2000). Once

bound to the cell, VacA is internalized by a pinocytic mechanism into GPI-AP-enriched early

endosomal compartments (GEECs) (Gauthier et al., 2007). Upon VacA’s internalization into

GEECs, it forms anion-selective channels by assembling into hexameric, ring like structures,

allowing chloride ions to pass through, resulting in swelling in endosomal membranes and

vacuole formation (Figure 1-2). These channels release bicarbonate and organic anions from the

cell cytosol to support bacterial growth (Cover and Blanke, 2005;Dhar et al., 2003;Szabo et al.,

1999). These large, intracellular vacuoles, with acidic intralumenal environments are enriched in

markers for late (but not early) endosomal compartments, such as Rab7, LAMP1 and Lgp110

(Papini et al., 1994).

Page 18: Ang Michelle 201006 MSc Thesis

5

Page 19: Ang Michelle 201006 MSc Thesis

6

Figure 1-1: H. pyori CagA signalling pathway.

CagA is injected into host cells upon H. pylori infection via the bacterial TFSS. Once injected,

CagA is localized near the cytoplasmic side of the plasma membrane until it is recruited by SRC

kineases. SRC kineases phosphorylate CagA on EPIYA motifs. Phosphorylated CagA then binds

SHP-2, activating SHP-2’s phosphatase activity and forming a complex that initiates mitogenic

signalling, cell scattering, elongation and rearrangement of the actin cytoskeleton.

Phosphorylation independent CagA interacts with c-Met and Grb2, as well as interacting with

ZO-1 and JAM to alter tight junctions.

Page 20: Ang Michelle 201006 MSc Thesis

Stomach Lumen

Oligomerization andMembrane interaction

Endocytic entry into cell

H+ (V-ATPase)

H2ONH3

NH4+

Cl- channel activity

Vacuole swelling

7

Page 21: Ang Michelle 201006 MSc Thesis

8

Figure 1-2: VacA induces cellular vacuolation.

Upon VacA release by H. pylori, VacA binds to the cell surface, where it oligomerizes and is

internalized via the GEEC pathway. Internalized VacA forms anion-selective channels which

allow chloride conductance, causing increased intraluminal chloride concentration. As a result,

increased ATPase activity occurs, increasing proton pumping which reduces intraluminal pH.

Membrane-permeant weak bases diffuse inwards and are trapped within these compartments.

Compartments undergo osmotic swelling, resulting in cellular vacuolation. Modified from Cover

and Blanke (2005)

Page 22: Ang Michelle 201006 MSc Thesis

9

The p33 fragment of VacA localizes specifically to the mitochondria where it affects

mitochondrial membrane permeability (Figure 1-3). VacA decreases the levels of 3, 3'-

dihexyloxacarbocyanine iodide, an indicator of mitochondrial membrane potential, followed by a

decrease in cellular ATP indicating mitochondrial and cellular damage (Kimura et al., 1999). The

decrease in mitochondrial membrane potential coincides with decreased ATP levels and O2

consumption which suggests that VacA damages the mitochondrial inner membrane (Kimura et

al., 1999). These mitochondrial effects are dependent on VacA’s ability to form anion-selective

channels described previously (Willhite and Blanke, 2004b). VacA also modulates the function

of several immune cells which will be discussed below.

In T-cells, VacA can inhibit T-cell proliferation and viability, by downregulating the IL-2 α-

receptor and inhibiting production of stimulatory IL-2 (Gebert et al., 2003;Sundrud et al., 2004).

VacA activates the MAP kinase pathway (Boncristiano et al., 2003;Sundrud et al., 2004) and the

NF-κB pathway in T-cells to promote proinflammatory effects through the induction of genes

involved in the classical NF- κB pathway (Takeshima et al., 2009). VacA inhibits the activation

of Jurkat T cells (a human T cell lymphoma/leukemia cell line) by inhibiting the activation of

NFAT, a key transcription factor required for the expression of genes involved in T cell

activation (Boncristiano et al., 2003;Gebert et al., 2003). Finally, it has been demonstrated that

VacA can suppress the proliferation of primary human CD8+ T cells and CD4

+ T-cells even

when strongly stimulated by potent T-cell activators (Torres et al., 2007).

VacA also acts on APCs, interfering with major histocompatibility complex (MHC) class II

mediated antigen presentation inhibiting the li-dependent pathway of antigen presentation. In

addition, VacA blocks antigen presentation on APCs to T-cells (Boncristiano et al.,

2003;Molinari et al., 1998).

B-cells have also been shown to be affected by the VacA toxin. VacA induces the apoptosis of

B-cells via the translocation of the apoptosis-inducing factor to the nucleus (Singh et al., 2006).

Similar to T-cells, VacA inhibits the activation and proliferation of B-cells, even when

stimulated by potent B-cell activators (Torres et al., 2007).

Page 23: Ang Michelle 201006 MSc Thesis

10

In macrophages, phagosome maturation is arrested by VacA, preventing the killing of H. pylori

by phagocytes (Zheng and Jones, 2003). In clinical isolates, there were lower observed levels of

bacterial multiplication in macrophages in strains that lacked the VacA gene (Wang et al., 2009).

There is some evidence to indicate that VacA has some pro-inflammatory effects, as increasing

levels of TNF-α and IL-6 due to VacA in mast cells was observed, however these effects need

further study (Cover and Blanke, 2005).

Page 24: Ang Michelle 201006 MSc Thesis

Surface bound VacA functioning as anadhesion

NucleusPro-in�ammatorysignalling

VacA

Localization tomitochondria

Cell death

Reduction of ∆ΨmRelease of cytochrome c

T-cell

Inhibition of T-cellactivation andproliferation

Vacuolation

Membranechannel activity

11

Page 25: Ang Michelle 201006 MSc Thesis

12

Figure 1-3: Multiple actions of H. pylori’s VacA toxin.

VacA toxin has many different effects within the cell. VacA can localize to the mitochondria,

causing a decrease in mitochondrial membrane potential, causing release of cytochrome c. VacA

can also be internalized and cause pro-inflammatory signalling, as well as interfere with the

activation and proliferation of T-cells. In addition, VacA induces formation of large intracellular

vacuoles as described previously. Modified from Cover and Blanke (2005).

Page 26: Ang Michelle 201006 MSc Thesis

13

1.3.3 Lipopolysaccharide (LPS)

Lipopolysaccharide (LPS) is an important structural component of the outer membrane of gram-

negative bacteria containing three parts: lipid A, a core oligosaccharide and an O side chain (Lu

et al., 2008). LPS contains pathogen-associated molecular patterns (PAMPs) which are

recognized by host cells to initiate innate responses. Lipid A is the main PAMP in LPS.

Normally, LPS stimulation of mammalian cells occurs through a series of interacting proteins

such as LPS binding protein (LBP), which facilitates interaction of LPS to CD14, a

glycosylphosphatidylinositol-anchored protein (Lu et al., 2008). CD14 interacts with Toll-like

receptor (TLR) 4, which upon LPS recognition, undergoes oligomerization, recruiting

downstream adaptors, initiating activation of the NF-κB signalling pathway, increasing

proinflammatory cytokine production through a myeloid differentiation primary response gene

88 (MyD88) dependant or independent pathway (Lu et al., 2008).

LPS from H. pylori possesses some unusual characteristics that differ from other gram-negative

bacteria’s LPS as it appears to be less potent at driving a pro-inflammatory response. H. pylori

LPS fails to induce inflammatory responses in epithelial cells at high concentrations (Backhed et

al., 2003). It is also known to be 1000 times less effective than E. coli LPS in inducing pro-

inflammatory cytokine production in macrophages/monocytes, as well as being less effective

than E. coli endotoxin in binding to LBP (Cunningham et al., 1996;Gobert et al., 2004). H.

pylori LPS signalling may also differ from the conventional pathway. It has also been reported

that TLR-2 not TLR-4 is the major TLR activated by LPS during H. pylori infection giving H.

pylori LPS a different signalling pathway from normal LPS which may attribute to its differing

characteristics (Smith, Jr. et al., 2003;Uno et al., 2007). Other studies suggest that H. pylori

strains act as antagonists for human TLR-4 (Lepper et al., 2005).

2 Innate Immunity

As H. pylori causes a chronic infection, the ability of the bacterium to evade host immune

responses including potentially escaping tumour immune surveillance is an area of great interest

and the focus of my thesis. The host immune system consists of both innate and adaptive

immunity. Innate immunity is a fast acting response, reacting quickly in bacterial infection with

the aim of eliminating the bacteria. The adaptive immune response is generally more delayed and

Page 27: Ang Michelle 201006 MSc Thesis

14

its responses are shaped by the innate immune response. An additional component of the host

immune response, which has recently been recognized, is the autophagy pathway. Below I will

summarize current knowledge with respect to the key areas of focus of my research in H. pylori

and innate immunity: dendritic cells and autophagy.

2.1 Dendritic Cells (DCs)

Dendritic cells (DCs) are a type of antigen presenting cell (APC) whose primary role is to

uptake, process and present antigens to T-cells, initiating and activating cytokine release and

immune responses (Lenahan and Avigan, 2006;Zou, 2005). Immature DCs are adept at antigen

capture, and circulate throughout the body until they encounter antigens. Following antigen

uptake, they process and convert proteins to peptides and present these processed peptides to T-

cells on MHC molecules (Steinman and Banchereau, 2007) (Figure 1-4). During this antigen

processing phase, these DCs mature and differentiate, increasing surface expression of MHC

class II, CD80 and CD86, which are costimulatory molecules necessary for T cell activation

(Lenahan and Avigan, 2006). Activated T-cells can have many responses whose direction is

dictated by APCs. Under the control of DCs, T-cells can acquire the capacity to produce

powerful cytokines to initiate a Th1 proinflammatory response, a Th2 antibody forming

response, a Th17 autoimmune response and even a T-regulatory (Treg) immunotolerance

response (Ferrero, 2005).

To stimulate T-cells, peptides must be processed by MHC molecules which will present the

peptide on the surface of the DC. The peptide-binding proteins are of two types, MHC class I

and MHC class II. MHC class I molecules assist in the processing of intracellular antigens and

will present them to cytotoxic T-cells which, once activated, can directly kill a target

(Banchereau and Steinman, 1998). MHC class II complexes recognize and process extracellular

antigens and present them to T-helper cells which can have profound immune-regulatory effects.

A normal inflammatory response initially consists of the recruitment of neutrophils, followed by

T cells, plasma cells, and APCs which is accompanied by epithelial cell damage (Goodwin et al.,

1986). The failure of the recruitment and maturation of DCs has been observed at tumour sites,

indicating that a dysfunctional DC response may lead to carcinogenesis (Lenahan and Avigan,

2006). Additionally it has been observed that there is an increase in the number of immature DCs

in patients with cancer, indicating that maturation is an important factor in DC function (Lenahan

Page 28: Ang Michelle 201006 MSc Thesis

15

and Avigan, 2006). In many cancer types the signal transducer and activator of transcription 3

(STAT3) pathway is upregulated, which has been shown to inhibit DC maturation for oncogenic

survival and proliferation (Lunz, III et al., 2007;Wang et al., 2004).

Page 29: Ang Michelle 201006 MSc Thesis

16

Page 30: Ang Michelle 201006 MSc Thesis

17

Figure 1-4: Model of DC activation.

Immature DCs circulating in the lamina propria come into contact with antigens released by

extracellular factors or the epithelial cell layer. Immature DCs sample these antigens and

increase their surface expression in many DC maturation markers such as CD86, CD80 and

MHC class II. Immature DCs are now mature DCs and APCs, where they present the processed

antigen to other immune cells such as T-cells to orchestrate the appropriate immune response.

Activation of the STAT3 pathway can inhibit the maturation process.

Page 31: Ang Michelle 201006 MSc Thesis

18

2.2 Signal Transducer and Activator of Transcription 3 (STAT3)

2.2.1 STAT3 Signalling Cascade

STAT3 is a transcription factor whose upregulation is thought to be involved in oncogenesis.

The STAT3 pathway can be activated by a number of different cytokines (such as IL-6), growth

factors and peptides, all signalling through gp130, a signal transducer receptor subunit that can

be activated by several members of the IL-6 family of cytokines (Hirano et al., 1997;Kim et al.,

2007a) (Figure 1-5). IL-6 activation of STAT3 occurs through the binding of the cytokine to its

receptor IL-6R, resulting in heterodimerization with the gp130 subunit (Kim et al., 2007a).This

heterodimer activates the receptor associated Janus Kinase (JAK) kinases which in turn

phosphorylate tyrosine residues on the cytoplasmic tail of the gp130 receptor, recruiting STAT3

to the phosphorylated residues (Greenlund et al., 1995). STAT3 monomers will then be

phosphorylated themselves on tyrosine 705 residues, and then dimerize through reciprocal Src-

homology-2 (SH2) domain phospho-tyrosine interactions. STAT3 dimers will be translocated

into the nucleus, using nuclear localization signals mediated by the importinα3/α6 complex,

which when interacting with importinβ allows the dimer to traffic through nuclear pore

complexes that span the nuclear membranes (Reich and Liu, 2006;Yang and Stark, 2008). Here,

central DNA binding domains located on the STAT3 monomers allow the dimer to bind directly

to response elements in the promoter regions of target genes to trigger induction of transcription

of several genes involved in growth and proliferation (Liu et al., 2005;Reich and Liu, 2006). The

STAT family of transcriptional regulators include an IFNα-stimulated gene response element

(ISRE) and the Gamma-IFN–stimulated activation sequence (GAS: TTTCCNGGAAA) (Horvath

and Darnell, 1997). This interaction induces the transcription of multiple genes involved in

apoptosis (surviving, Bcl-nL, HSP27), cell cycle regulation (cyclin D1, c-myc, c-fos),

angiogenesis (vascular endothelial growth factor), cell invasion, metastasis and immune function

(Kim et al., 2007a;Kisseleva et al., 2002;Levy and Darnell, Jr., 2002). STAT3 dimers can also be

phosphorylated on the serine 727 residue. Phosphorylation of this residue results in dimers with

increased transcriptional activity for recognition sites involved in the mitogen-activated protein

(MAP) kinase pathway (Horvath and Darnell, 1997).

Page 32: Ang Michelle 201006 MSc Thesis

19

Page 33: Ang Michelle 201006 MSc Thesis

20

Figure 1-5: The STAT3 activation pathway.

IL-6 molecules bind to the IL-6-αR. This leads to heterodimerization with the gp130 subunit.

This dimer activates associated JAK kinases which phosphorylate residues on the cytoplasmic

tail of the gp130 receptor. Cytoplasmic STAT3 monomers are recruited to these phosphorylated

sites and form dimers via their reciprocal SH2 phospho-tyrosine interactions. Dimeric STAT3

translocates to the nucleus via importins-α3 and α6 through nuclear pore complexes and leads to

the transcription of genes involved in survival, proliferation and angiogenesis, potentially leading

to carcinogenesis.

Page 34: Ang Michelle 201006 MSc Thesis

21

2.2.2 STAT3, DCs and Cancer

Evidence suggests that STAT3 is involved in carcinogenesis, as this pathway is constitutively

activated in 50-90% of human cancers (Kortylewski and Yu, 2007). Disrupting constitutive

STAT3 activation in tumours results in the induction of apoptosis by increasing proinflammatory

cytokine and chemokine expression, indicating that STAT3 activation may play a role in the

reduction of proinflammatory responses, and thus increase immune evasion (Wang et al., 2004).

Inhibition of STAT3 results in a production of inflammatory mediators that can activate DCs

(Wang et al., 2004). Additionally, disruption of STAT3 activates innate immune cells in vivo,

and also effects adaptive immunity from DCs as when STAT3 is activated, DCs do not undergo

maturation (Wang et al., 2004). Similarly, blocking STAT3 in tumours leads to the activation of

T cells, promoting DC maturation, demonstrating the ability of STAT3 to inhibit DC mediated

immune responses (Wang et al., 2004).

2.2.3 STAT3 and H. pylori

We have shown that H. pylori activates the STAT3 pathway in gastric epithelial cells in vitro and

in vivo in a CagA dependant mechanism, thereby favoring the development of gastric cancer

(Bronte-Tinkew et al., 2009). In the Mongolian gerbil, STAT3 activation was detected in gastric

epithelial cells upon infection with CagA+ H. pylori (Bronte-Tinkew et al., 2009). In addition,

STAT3 activation was detected in infiltrating immune cells during infection of Mongolian

gerbils (Figure 1-6) suggesting that activation of this pathway may alter host immune responses.

2.3 Dendritic Cells and H. pylori

As DCs are key modulators of immune responses, the ability of H. pylori to alter DC function is

of great interest. How H. pylori alters DC function is controversial. DCs are capable of entering

H. pylori infected gastric epithelium, where they uptake bacteria directly through the extension

of long luminal projections by DCs through epithelial apical-junctions (Necchi et al.,

2009;Wilson and Crabtree, 2007) (Figure 1-7). H. pylori induces the secretion of many different

cytokines such as IL-6, IL-8, IL-10, IL-12, IL-1, and tumour necrosis factor (TNF) (Kranzer et

al., 2005;Rad et al., 2007).

Page 35: Ang Michelle 201006 MSc Thesis

Control

H. pylori infected

22

Page 36: Ang Michelle 201006 MSc Thesis

23

Figure 1-6: From Bronte-Tinkew et al. Nuclear STAT3 detected in epithelial and

inflammatory cells of H. pylori CagA+ infected Mongolian gerbil tissue.

A) Mongolian gerbil stomach sections were stained via immunohistochemistry with STAT3

(red) and DAPI (blue) antibodies. Nuclear STAT3 is detected in the inflammatory cell infiltrate

from H. pylori infected stomach sections from gerbils (red) (Bronte-Tinkew et al., 2009).

Page 37: Ang Michelle 201006 MSc Thesis

Ureaseother HP factors

IL-12IL-10

IL-23

IL-10TGF-β

IL-10TGF-β

IL-4IL-5IL-10

Th1

Th2

Th17

Treg

B-cell

IL-12

IFN-γTNF-αIL-2

24

Page 38: Ang Michelle 201006 MSc Thesis

25

Figure 1-7: Summary of the possible immune responses to H. pylori. Dashed lines represent

the components of the pathway that are more speculative. H. pylori has several effects on the

immune response, effecting the release of several cytokines that alter Th1, Th2 and Treg

responses. Modified from Wilson and Crabtree (2007).

Page 39: Ang Michelle 201006 MSc Thesis

26

The effects of this cytokine secretion are unclear. Some studies demonstrate that H. pylori can

induce DC maturation, upregulating DC surface expression of CD80, CD83 and CD86 (Kranzer

et al., 2004;Rad et al., 2007). Conversely, other studies show that H. pylori inhibits DC

activation via modulation of IL-12 secretion, potentially leading to immune evasion (Kao et al.,

2006;Obonyo et al., 2006). In our studies with the Mongolian gerbil, activation of STAT3 was

detected in infiltrating immune cells. Therefore, I hypothesized that H. pylori infection would

activate STAT3 in dendritic cells, thereby altering host immune responses. Determining the

effect of H. pylori infection on STAT3 activation in dendritic cells is the focus of chapter three

of my thesis.

2.4 Autophagy

Another important host response which modulates immunity and infection is autophagy.

Autophagy is a process that involves the sequestering and transit of cytoplasmic components or

organelles to the lysosome for degradation. These broken down substrates are then used to

produce amino acids for protein and energy synthesis (Meijer and Codogno, 2006). There are

three types of autophagy: Microautophagy, macroautophagy and chaperone-mediated autophagy.

Microautophagy is responsible for engulfing large structures, sequestering cytosolic components

directly by lysosomes through invaginations in their membranes (Klionsky and Emr, 2000).

Chaperone-mediated autophagy can only degrade soluble proteins, and involves direct

translocation of unfolded substrate proteins across the lysosome membrane (Klionsky and Emr,

2000). Macroautophagy (henceforth referred to as autophagy), sequesters and engulf large

structures within a unique double-membraned vesicle in selective and non-selective processes

(Klionsky and Emr, 2000). Subsequently, these cargo are delivered to the lysosome where the

cargo is degraded or recycled (Levine and Klionsky, 2004).

Upon induction of autophagy, the process begins with the formation of double-membrane

compartments called autophagosomes. The origin of these membrane compartments is unclear. It

is thought that membrane compartments may originate from the ER, however evidence also

demonstrates that the pre-autophagosomal structure (PAS) contains Atg9, and in addition to

localizing with the PAS, membrane components can also be visualized with Mitoflour, a dye

used to identify mitochondrial membrane components, suggesting that the mitochondria may

provide components for the autophagosome (Klionsky and Emr, 2000;Reggiori et al., 2005).

Page 40: Ang Michelle 201006 MSc Thesis

27

The formation of the PAS and autophagosome involves several Atg genes, with Atg9 being the

only transmembrane protein in the core machinery that is conserved among species (Xie and

Klionsky, 2007) (Figure 1-8). Two ubiquitin-like conjugation reactions are needed to form the

Atg12-Atg5 complex. The carboxy COOH terminal of Atg12 is activated by linking first to

Atg7, an E1-like enzyme, which passes the Atg12 to Atg10, an E2-like enzyme, which is then

attached to the internal lysine residue 130 of Atg5, later recruiting Atg16 to form an Atg12-Atg5-

Atg16 multimer complex (Klionsky and Emr, 2000). This complex is present on the PAS,

however dissipates upon autophagosome formation. Autophagosomal elongation is mediated by

mictotubule-associated light chain (LC)-3 (Atg8 in yeast). The second reaction of LC3 lipidation

which joins LC3 to phosphatidylethanolamine (PE) first involves Atg4 which cleaves LC3’s

carboxy terminal leaving a conserved glycine residue (Klionsky and Emr, 2000). Cleaved LC3

next binds to Atg7, then Atg3 and finally to autophagosomal membrane protein PE, where it

becomes membrane-bound LC3-II (Klionsky and Emr, 2000). This complex is present in the

initial stage of autophagosome formation, and remains on the autophagosome throughout

formation and fusion with the lysosome and is often used as a marker of autophagy in

experimentation (Yorimitsu and Klionsky, 2005).

Complete autophagosomes are then delivered to lysosomes in a manner dependant on

microtubules and maintenance of proper acidification(Klionsky and Emr, 2000). Intermediate

autophagosomes fuse with lysosomes and acquire cathepsins and acid phosphatases to become

mature autolysosomes, where the inner autophagosomal membrane is degraded.

Autophagy plays an important role in immunity, as autophagic machinery is often used in

defence against microbes, delivering microorganisms to lysosomes for degradation (a process

called xenophagy) and the delivery of microbial nucleic acids and antigens to

endosomal/lysosomal compartments to activate innate or adaptive immunity (Levine and

Kroemer, 2008). Several bacteria are degraded by xenophagy, including Mycobacterium

tubercuolosis, Shigella flexneri, Salmonella enterica, and Listeria monocytogenes (Huang and

Klionsky, 2007;Levine and Kroemer, 2008). Atg5 has been shown to be crucial in the delivery of

viral nucleic acids from the Sendat virus to endosomal TLR-7 receptors which then activate DCs

(Lee et al., 2007). Autophagy is also involved in the delivery of synthesized microbial antigens

to MHC class II APCs which subsequently activate CD4+ T cells (Paludan et al., 2005).

Page 41: Ang Michelle 201006 MSc Thesis

Formation ofSequestration Cresent

Lysosome

Lysosomal Hydrolases

Docking& Fusion

Atg5-Atg12-Atg16 complex

LC3 II

28

Page 42: Ang Michelle 201006 MSc Thesis

29

Figure 1-8: Overview of the autophagic process in mammalian cells. Cytosolic components

and organelles are either specifically or randomly targeted by the PAS. A sequesteration crescent

forms around these components initially requiring the Atg5-Atg12-Atg16 complex (red dots),

followed by LC3 lipidation into the LC3II form (yellow dots). The Atg5-Atg12-Atg16 complex

disassociates prior to autophagosome completion, while LC3II remains on the autophagosome

throughout. Lysosomes are trafficked to completed autophagosomes, where they fuse. The inner

membrane of the autophagosome is degraded, followed by degradation of autophagosomal

contents by lysosomal hydrolases.

Page 43: Ang Michelle 201006 MSc Thesis

30

Autophagy also has been shown to have a role in tumourigenesis as failure to clear the

intracellular debris normally eliminated by autophagy can lead to tissue damage, cell death and

chronic inflammation that is tumour promoting (White et al., 2010). Atg6/beclin1 is a

haploinsufficient tumour suppressor in mice (Qu et al., 2003;Yue et al., 2003). Atg6/beclin1 has

been shown to be monoalleically deleted in ovarian and breast cancer (Qu et al., 2003).

Inactivating mutations in Atg5 were found to be infrequently present in colorectal cancer (Jung

et al., 2008). Losses of other autophagy mediators such as p63 and Atg4c were also shown to

enhance tumour formation (Marino et al., 2007;Mathew et al., 2009).

Basal levels of autophagy are responsible for the clearance of damaged organelles, such as the

mitochondria. The importance of mitochondrial degradation is expected, given the fact that

mitochondrial homeostasis in a cell is crucial. Specific autophagy targeting the mitochondria

(mitophagy) has been a growing field of interest, as mitochondrial instability, such as changes in

Mitochondrial Permeability Transition (MPT) can play a role in the initiation of mitophagy.

Mitochondrial production of ROS is an important factor for the overall ROS level within a cell.

Mitochondria ROS production produces superoxide and H2O2 through its respiratory chain, and

accumulation of these ROS can lead to lipid peroxidation, apoptosis and necrosis, mitochondrial

dysfunction and DNA damage (Murphy, 2009). Additionally, the ROS produced by the

mitochondria have been reported to regulate autophagy and autophagic cell death, as autophagy

is induced by rotenone, H2O2, and superoxide (Chen and Gibson, 2008). ROS production can

also lead to 10-20 fold higher mutation rates in mitochondrial DNA versus nuclear DNA (Yakes

and Van Houten, 1997) which may also induce mitophagy. Failure to clear the cell of damaged

mitochondria can lead to cellular damage, thus leading to cell death (Kim et al., 2007b).

Oxidative damage due to ROS production plays an important role in tumourigenesis as increased

ROS levels are often associated with cancerous cells. Increased ROS levels can lead to DNA

damage, implicated in mutating tumour suppressor p53 (Morgan et al., 2003) causing

carcinogenesis. Additionally, ROS production is involved in Ras-Raf-MEK signalling. This

pathway is important in mediating protection against apoptotic cell death induced by increased

oxidative stress, directly enhancing the activation of Ras, and augmenting ERK1/2, leading to

oncogenesis (Pan et al., 2009). Lipid peroxidation leading to ROS production has also been

shown to cause DNA damage due to the production of high levels of oxygen radicals (Marnett,

Page 44: Ang Michelle 201006 MSc Thesis

31

2000). Thus there is a well-established link between ROS generation and tumourigenesis which

may prove to play a role in H. pylori mediated tumourigenesis.

2.4.1 H. pylori and Autophagy

Recently, our lab has shown that H. pylori can induce autophagy in human gastric epithelial

(AGS) cells, and that its VacA toxin is sufficient to induce this autophagy (Terebiznik et al.,

2009). Induction of autophagy was dependant on VacA’s channel-forming ability, and limited

the effects of VacA (Terebiznik et al., 2009). However, the exact mechanisms by which VacA

induces autophagy remain unknown. I hypothesized that VacA mediated damage to

mitochondria may be sufficient to induce autophagy. Determining the mechanisms responsible

for VacA mediated autophagy is the focus of chapter four of my thesis.

3 Summary

In summary H. pylori evades host responses to cause a chronic infection and promote

carcinogenesis in a subpopulation of those infected. Despite intense research in the field, the

mechanisms by which H. pylori evades the host immune response are still unclear. In my thesis,

I will explore two distinct avenues by which H. pylori may alter or hide from the innate immune

response. Firstly, I will explore the interaction of H. pylori on a key orchestrator of the immune

response: the dendritic cell. Through interactions with this key APC, H. pylori may significantly

alter the outcome of the innate immune response. Secondly, I will explore the role of H. pylori

mediated autophagy to try and determine the mechanisms responsible for the induction of this

response. Changes in the autophagic pathway may modify the host cell immune response,

providing another mechanism by with H. pylori alters the innate immune response.

Overall hypothesis

The overall hypothesis of my thesis is that H. pylori effects host innate immune responses to

promote chronic infection and disease.

Specific aims

The specific aims of my thesis are:

Page 45: Ang Michelle 201006 MSc Thesis

32

1. To determine whether H. pylori activates the STAT3 pathway in DCs thus altering the host

immune response and providing a mechanism for H. pylori’s immune evasion

2. To determine the mechanisms by which H. pylori’s VacA toxin induces autophagy

Page 46: Ang Michelle 201006 MSc Thesis

33

Chapter 2 Materials and Methods

CHAPTER 3 MATERIALS AND METHODS

Bone Marrow Derived DCs

Bone marrow-derived DCs (BMDCs) were obtained and cultured from wild-type (WT) C57BL/6

mice. Bone marrow was extracted from the femur and tibia and seeded at 2 x 106/plate in

bacteriological petri dishes with 10ml RPMI (Roswell Park Memorial Institute)-1640 with L-

glutamine and 25mM HEPES supplemented with 1X nonessential amino acids, 50 mM 2-β-

mercaptoethanol, 1mM Sodium pyruvate, 100U/ml of penicillin/streptomycin (all from

Invitrogen, Burlington, Ontario, Canada), and 10% fetal bovine serum (FBS) (Gibco BRL Life

Technologies, Gaithersburg, MD, USA) supplemented with 100 ng/ml of granulocyte-

macrophage colony-stimulating factor (GM-CSF). Cells were incubated at 37°C for 3 days

before an additional 10ml of RPMI mixture was added. On day 6, cells were harvested and

magnetically sorted for the CD11c+ surface marker (Miltenyi Biotec, Bergisch Gladbach,

Germany) to ensure a pure DC population. Sorted cells were then plated in 6-well tissue culture

plates and used for stimulation.

H. pylori Strains and Infection Conditions

H. pylori gerbil adapted strain 7.13 or the corresponding isogenic cagA- mutant were grown on

blood agar plates containing 5% sheep blood under microaerophilic conditions (5% O2, 10%

CO2, 85% N2). Isogenic mutant strains were grown on Columbia blood agar plates supplemented

with 10% FBS and 20μg/ml kanamycin under similar conditions. After a monolayer of bacteria

were grown on the plates, bacteria were then transferred and grown in Brucella broth with 10%

FBS for 18 h, harvested by centrifugation, and added to gastric cells at a multiplicities of

infection (MOI) of 25:1. In some experiments, H. pylori was heat killed by boiling for 20 min in

sealed sterile Eppendorf tubes. Heat killing was confirmed by lack of growth on Columbia blood

agar plates. Where indicated, 30 min before infection, cells were incubated with the STAT3

inhibitor S3I-201at 25μM (Calbiochem, La Jolla, CA, USA). For controls, cells were incubated

with IL-6 (50ng/ml; Cell Signalling, Danvers, MA, USA) for 30 min, or 24 hours; or E. coli LPS

(1μg/ml; Sigma Aldrich) for 24h.

Page 47: Ang Michelle 201006 MSc Thesis

34

Cell Lysates and Western Blotting

After 4h or 24h of infection, cells were washed and scraped in 75μl of RIPA buffer containing

phosphatase and protease inhibitors (all from Sigma Aldrich). Cell suspensions were centrifuged

and supernatants stored at -80°C. Protein concentrations were measured using the Biorad Dc

protein assay. Lysates in laemelli buffer were loaded onto 10% SDS-polyacrylamide separating

gels and run at 110V for 1.5h at room temperature. Proteins were then transferred onto a

nitrocellulose membrane (BioTrace NT; Pall Corp., All Arbor, MI, USA) at 100V for 1.5 hours

at 4°C. Membranes were blocked with Tris-buffered saline with tween (TBST), 5% skim milk

followed by incubation with either rabbit anti-phospho-STAT3 antibody (1:250; Cell Signalling

MA, USA) or mouse anti-STAT3 antibody (1:500, Cell Signalling) in TBST milk. Blots were

then incubated with corresponding secondary horseradish peroxidase-conjugated antibody

(Jackson Laboratories, PA, USA) for 1h at room temperature. Bands were visualized by

chemiluminescence using Kodak Biomax MR film.

Densitometric analysis

Densitometric analysis was performed using FlourChem FCII software. All blots from each

independent experiment were used. Densities of phosphorylated STAT3 (PSTAT3) and STAT3

bands were measured for each treatment and expressed as a ratio (PSTAT3/STAT3).

Fluorescence Activated Cell Sorting (FACS) – Flow Cytometry

BMDCs infected for 24h were utilized for flow-cytometric analysis. Cells were scraped gently

on ice in PBS and spun at 350g for 10 min. Cells were resuspended in FACS buffer (PBS

containing 0.05% of sodium azide). Cells were incubated with CD16/32 (Fc receptor block)

(1μl/106 cells; eBioscience, San Diego CA, USA) for 15 minutes at 4°C prior to staining to block

nonspecific staining. Subsequently, cells were washed with FACS buffer and incubated with the

following antibodies: Flourescein isothiocyanate (FITC)-labeled antibody to CD11c,

phycoerythrin (PE)-labeled antibodies to CD86 and CD80, peridinin chlorophyll protein-cyanine

(PerCP-Cy5.5)-conjugated antibody to CD11b (all from eBioscience), biotin-labeled antibodies

specific for MHC class II followed by streptavidin conjugated to allophycocyanin (APC) (Becton

Dickson, San Jose, CA, USA). After staining, cells were washed twice with FACS buffer and

Page 48: Ang Michelle 201006 MSc Thesis

35

analyzed by FACS (FACSCaliburflow cytometer; Becton Dickinson). FlowJo software was used

for the analysis of the results.

ELISA Cytokine Analysis

BMDCs were infected for 24h as described above. Supernatants were collected and flash frozen

and stored at -80°C. Supernatants were assayed for IL-6, IL-10, IL-12p40 and TNF-α using

specific ELISAs as per the manufacturer’s instructions (R&D systems, MN, USA).

Multiplex Bead-based Luminex Assay

Supernatants from BMDCs were assayed for IL-2, IL-4, IL-5, IL-10, IL-12, IL-17 and IFN-γ

using the Th1/Th2 Cytokine Mouse 6-Plex Panel kit plus the IL-17 Mouse Singleplex Bead

Luminex Assay Kit as per the manufacturer’s instructions (Invitrogen).

MTT live/dead cell Assay

To determine whether cell viability was affected by the addition of S3I-201 or other agents, an in

vitro toxicology MTT assay (Sigma Aldrich) was performed. BMDCs were grown and infected

for 24h as described. Viable cells contain mitochondrial dehydrogenase, which cleaves the

tetrazolium substrate ring, yielding purple formazan crystals. The degree of mitochondrial

enzymatic activity was determined via the dissolution of formazan precipitate and

spectrophotometric analysis of the resulting solutions from each treatment.

CHAPTER 4 MATERIALS AND METHODS

Cell and Bacterial Culture

Human gastric epithelial cells (AGS) were grown in Ham’s F-12 media (Wisent St. Bruno, PQ,

Canada) supplemented with 10% FBS and incubated at 37°C in a CO2 environment.

H. pylori strain 60190 from frozen stock was inoculated on blood agar plates with 5% sheep

blood (Oxoid Biotech, Canada). Isogenic mutants lacking the vacA gene were inoculated on

Brucella agar plates supplemented with 10% fetal bovine serum (FBS) (Gibco BRL Life

technologies) and 20μg/ml kanamycin (Invitrogen). All cultures were incubated at 37°C in a

microaerophilic environment for 72h. Cultured H. pylori strains were then grown at 37°C

Page 49: Ang Michelle 201006 MSc Thesis

36

overnight in Brucella broth (Sigma-Aldrich) under microaerophilic conditions with shaking at

120 rpm. On the day of infection, bacteria were pelleted and resuspended in Ham’s F12 medium

with 10% serum.

Preparation of Concentrated Culture Supernatants

The culture supernatants of wt H. pylori 60190 and isogenic mutant vacA- were separated from

the bacterial pellet at 4000rpm for 20 min. Supernatants were then filtered through a 0.22μm

filter and concentrated 10 times in Ham’s F12 media supplemented with 10% FBS using a 30

kDa cut off Amicon Ultra centrifugal filter (Millipore, Billerica, MA, USA). The culture

supernatants are diluted a further 5 times in Ham’s F12 prior to addition to cells for intoxication

assays (Raju and Jones, 2010).

Purified VacA Toxin

VacA toxin was purified by chromatography (Willhite et al., 2003) (obtained from Dr. Steven

Blanke, University of Houston, Houston, Texas USA) and diluted 100 times in Hams F12,

adjusted to pH 2.0 using 1-Normal HCl to acid activate the toxin. Purified toxin was left at 37°C

for 30 min followed by neutralization using 1-Normal NaOH. Toxin is supplemented with 10%

FBS. Prior to addition of activated toxin to cells NH4Cl was added (5mM).

Antioxidants and Chemicals

Where indicated, cells were pre-treated with several antioxidants for 30 minutes. The following

antioxidants were used: Rotenone (50μM), α-tocopherol (100μM), diphenylene iodonium (DPI;

100 μM), n-acetylcysteine (NAC; 5mM) (all courtesy of the John Brumell’s laboratory,

University of Toronto, Toronto, Ontario, Canada), and Tempol (5mM; Sigma Aldrich).

Autophagy Inducers

Autophagy was induced by incubating AGS cells in Earle’s balanced salts solution (EBSS,

Gibco) or with rapamycin (25µg/ml) at 37°C for 4h.

Infection Conditions

Page 50: Ang Michelle 201006 MSc Thesis

37

Infection of AGS cells was performed in 6-well plates when 80-90% confluency was reached.

Cells were treated with culture supernatants, purified VacA toxin or inactive VacA toxin for 4h.

Where indicated, pretreatment with antioxidants was used for 30 min, followed by subsequent

VacA treatments. Upon completion, cells were either lysed for western blotting.

Immunoflourescence

AGS cells were grown on coverslips or 12-24-well plates. Cells were transfected using

FuGENE-6 (Roche Diagnostics Indianapolis, Indiana, USA) as a transfection agent for various

plasmids. Plasmids LC3-GFP, LC3-RFP (Obtained from John Brumell’s lab) Mito-RFP, and

Parkin-YFP (both kindly provided by Dr. Peter Kim, University of Toronto, Toronto, ON,

Canada) were used alone, or in combination as needed. Upon infection completion, cells were

washed with PBS and fixed in 4% paraformaldehyde for 20 min. Cells were then permeabilized

by incubation in 0.1% Triton X-100 (vol/vol) in PBS for 20 min, and blocked for 60 min with

5% milk in PBS (vol/vol). Permeabilized cells were incubated for 1 h with primary antibody at

room temperature, washed extensively with PBS buffer, and incubated for 1 h with secondary

antibodies. All steps were carried out at room temperature. The following primary antibodies

were diluted in 5% milk in PBS (vol/vol) as follows: HSP60 (1:40; Abcam, Cambridge, MA,

USA), and VacA (1:100; from the Blanke Lab), followed by washing in PBS and a Cy3 or Cy5

conjugated secondary antibody (dilution 1:1000). After washing in PBS, the cover slips are then

mounted onto microscope slides.

Parkin

Experiments examining Parkin used Parkin-YFP-transfected cells treated with VacA pure toxin

as described above. Rotenone (50µM) was used as positive control for Parkin redistribution.

Confocal Microscopy

Confocal image acquisition was performed in a Quorum Spinning Disk Confocal Microscope

consisting of a Leica DMIRE2 inverted fluorescence microscope equipped with a Hamamatsu

Back-Thinned EM-CCD camera and spinning disk confocal scan head, 4 laser lines (Spectral

Applied Research: 405 nm, 491 nm, 561 nm, 638 nm), an ASI motorized XY stage, and an

Improvision Piezo Focus Drive. The equipment is controlled by Volocity acquisition software

Page 51: Ang Michelle 201006 MSc Thesis

38

(Improvision) and powered by an Apple Power Mac G5 (Terebiznik et al., 2009). The number of

autophagic cells was enumerated in approximately 100 LC3-GFP transfected cells using the 63X

objective. Cells containing more than five visible autophagosomes were considered autophagic.

Quantification of Parkin redistribution was also counted in approximately 100 Parkin-YFP

transfected cells using the 63X objective. Cells containing visible Parkin puncta were counted as

Parkin-redistributed.

Immunoblotting

AGS cells in 6 well plates were treated for 4 h. Western blotting technique was similar as that

used previously in the chapter 3 methods, using GFP polyclonal antibody (1:1000) with a

secondary HRP-conjugated anti-rabbit antibody for detection of LC3-GFP.

Reactive Oxygen Species

AGS cells were grown in 12-well plates and treated for 4 h as described. Cells were then washed

in PBS 2X, and 300μl of 0.25% trypsin (Wisnet) was added until cells detached. Trypsin was

neutralized with 300μl Ham’s F12. Cells were spun at 2500rpm for 10min at room temperature.

Cells were washed with buffer containing PBS plus 1% bovine serum albumin (BSA; Sigma

Aldrich) and 0.05% azide. Cells were then incubated with 20mM 5-(and-6)-chloromethyl-2'7'-

dichlorodihydrofluorescein diacetate acetyl ester (CM-H2DCFDA; Invitrogen), a dye that

remains nonfluorescent until the acetate groups are removed by intracellular esterases and

oxidation occurs within the cell. Radical formation was assessed via flow cytometry using a

FacsCalibur flow cytometer.

Statistics

All experiments were performed at least 3 times, and using values obtained, P values were

calculated using ANOVA to compare the means ± SE for independent treatment groups. A P

value of less than 0.05 was determined to be statistically significant.

Page 52: Ang Michelle 201006 MSc Thesis

39

Chapter 3 H. pylori Activates the STAT3 Pathway in DCs, Causing DC Maturation

but Altering DC Cytokine Release

ABSTRACT

H. pylori colonizes the stomach of nearly half the world’s human population and is an important

risk factor in the development of gastric cancer. Infection with H. pylori persists for the lifetime

of the host, utilizing mechanisms to evade the host immune response. The effect of H. pylori

infection on DCs, key orchestrators of the host immune response, is unclear. Recently, our lab

demonstrated that H. pylori activates the STAT3 pathway in epithelial cells. STAT3 activation in

DCs effects maturation, activation and can subvert tumour immune surveillance. Therefore, we

hypothesized that H. pylori activates the STAT3 pathway in DCs, altering maturation, thereby

providing a mechanism by which this bacteria evades host immune responses. BMDCs were

infected with H. pylori for 4 or 24 hours and activation of STAT3 was assessed by

immunoblotting. DC activation and maturation was assessed by flow cytometric analysis and

cytokine analysis. Infection of DCs with WT H. pylori resulted in robust PSTAT3 activation

compared to control, however this activation was independent of CagA. Activation of PSTAT3

was also seen with heat-killed wild-type bacteria, indicating a possible role for heat stable LPS.

STAT3 activation was associated with an induction of DC maturation markers and a cytokine

profile of increased IL-10: IL-12, providing a possible mechanism by which immune evasion

occurs. These findings suggest that H. pylori mediated STAT3 activation in DCs promotes an

immunoregulatory phenotype which may promote chronic infection and disease.

INTRODUCTION

Gastric cancer is the second leading cause of cancer-related deaths worldwide (Peek, Jr. and

Crabtree, 2006). As a type I carcinogen, infection with H. pylori is a leading cause of gastric

cancer and due to its long term and chronic infection, likely alters host immune response for

bacterial survival.

Dendritic cells are key players that orchestrate host immune responses. DCs are antigen

presenting cells that detect infection through recognition of pathogen associated molecules to

either induce or prevent the initiation of adaptive immune responses. In addition to responding to

Page 53: Ang Michelle 201006 MSc Thesis

40

infection, DCs are also involved in tumour surveillance. Classically immature DCs were thought

to promote tolerance while mature DCs promote T cell immunity. However, current evidence

suggests that DCs can exist in a variety of functional forms with different immunogenic

capacities (Banchereau and Steinman, 1998;Tan and O'Neill, 2005).

Based on their ability to regulate both host immune responses and tumour surveillance, the effect

of H. pylori infection on DCs has been an area of interest. However, controversy exists with

respect to the effect of H. pylori infection on DC function. Several studies suggest that H. pylori

can induce DC maturation, upregulating CD80, CD83 and CD86 surface expression by an as yet

unknown mechanism (Kranzer et al., 2004;Rad et al., 2007). The functional outcome of this

maturation is unclear. Some groups report an increase in DC IL-12 secretion (Guiney et al.,

2003) and a Th1 response, whereas other groups demonstrate inhibition of DC IL-12 secretion

when compared to other bacteria (Kao et al., 2006;Obonyo et al., 2006). Differences in cell lines

could contribute to the differences noted by these groups, as Guiney et al. performed

experiments on human peripheral blood monocytes treated with IL-4, which preferentially

polarizes cells toward an IL-12/Th1 response (Guiney et al., 2003), whereas Kao and Obonyo

used BMDCs cultured from mice with IL-4 treatment (Kao et al., 2006;Obonyo et al., 2006).

Conversely, others have demonstrated that H. pylori causes gastric epithelial cells to promote

thymic stromal lymphopoietin causing increased DC production of IL-4 and IL-13, causing a

Th2 response (Kido et al., 2010). Recently, it has been suggested that H. pylori induces DCs to

secrete factors causing a Treg polarized response through secretions of IL-10 and TGFβ (Kao et

al., 2009;Romero-Adrian et al., 2010).

Our laboratory has shown that H. pylori activates the STAT3 pathway in epithelial cells (Bronte-

Tinkew et al., 2009). STAT3 is constitutively activated in many cancer cells but is also increased

in infiltrating immune cells. Recent evidence indicates that STAT3 can suppress anti-tumour

immunity by inhibiting proinflammatory cytokine secretion and upregulating

immunosuppressive cytokines such as IL-10 and VEGF. STAT3 activation in DCs has been

shown to inhibit maturation, as blocking STAT3 increases expression of surface markers MHC

class II and CD40 (Wang et al., 2004). Blocking STAT3 also induces increased IL-12, IL-10,

TNFα and IL-6 promoting the development of Treg cells (Wang et al., 2004). Additionally, in

liver DCs, constitutively elevated IL-6 and STAT3 levels help inhibit liver DC maturation,

preventing robust inflammation in response to commensal bacteria (Lunz, III et al., 2007). These

Page 54: Ang Michelle 201006 MSc Thesis

41

studies suggest that, STAT3 contributes to the inhibition of DC maturation, thereby dampening

the initiation of robust immune responses by DCs. Therefore, with this body of evidence

implicating STAT3 as a modulator of DC function, we wanted to determine if the STAT3

pathway is activated during H. pylori infection and what effects it has on the DC phenotype.

RESULTS

H. pylori infection induces tyrosine phosphorylation of STAT3

To determine if H. pylori activates the STAT3 pathway in DCs, we measured phosphorylated

STAT3 (PSTAT3) in cell lysates using immunoblotting. Cultured immature BMDCs were

infected with H. pylori WT strain 7.13 or treated with IL-6, a known STAT3 activator, for 4h.

Immunoblotting analysis of lysates demonstrated robust activation of PSTAT3 with WT H.

pylori infection and IL-6 treatment in comparison with uninfected control cells (Figure 3-1A,

C). As we had previously shown that CagA is responsible for STAT3 activation in epithelial

cells we next determined if CagA was involved by utilizing the isogenic cagA mutant. Infection

with cagA- bacteria increased PSTAT3 in a manner similar to WT (Figure 3-1A) indicating that

phosphorylation of STAT3 is independent of the CagA virulence factor. Next we determined if

heat-stable proteins were required by employing heat killed (HK) bacteria. In comparison with

control cells, lysates from cells infected with HK bacteria showed increased PSTAT3 (Figure 3-

1B). However the degree of PSTAT3 was decreased in comparison with infection with the

wildtype strain, indicating that although heat-stable factors may contribute to STAT3 activation,

live WT bacteria are needed for full PSTAT3 activation.

Page 55: Ang Michelle 201006 MSc Thesis

Cont

rol

IL-6

WT

7.13

cagA

- 7.1

3

PSTAT3

STAT3

Cont

rol

IL-6

WT

7.13

HK 7

.13

PSTAT3

STAT3

* P < 0.05

A

B

C

42

Page 56: Ang Michelle 201006 MSc Thesis

43

Figure 3-1: H. pylori activates the STAT3 pathway in DCs.

A) Lysates from DCs incubated with IL-6 (100ng/ml, 30 min) or infected with H. pylori WT

strain 7.13, isogenic mutant cagA- or B) HK bacteria (MOI 25:1) for 4h were used for

immunoblotting to detect changes in PSTAT3 . C) Graph represents densitometric analysis of

PSTAT3 bands obtained for each protein signal normalized to native STAT3 bands. Fold

increase is given as a ratio of control PSTAT3/STAT3. Columns, mean; bars, SE, *, P<0.05,

using one-way ANOVA (n=6).

Page 57: Ang Michelle 201006 MSc Thesis

44

H. pylori induces expression of DC maturation markers

We next determined the effect of H. pylori infection on maturation of DCs. BMDCs were

infected with WT, HK H. pylori or incubated with IL-6 or E. coli LPS (positive control) for 24h.

Flow cytometric analysis was utilized to quantify expression of DC maturation markers CD86,

CD80, and MHC class II. Cells positive for both MHC class II and one other marker were

determined to be activated and mature. To ensure that a pure population of DCs was used, cells

were stained and gated for the CD11c+ and CD11b

+ surface markers (Figure 3-2A). Cells were

also stained for Gr1 to ensure the cells were not granulocytes (data not shown). Infection with

WT H. pylori increased the proportion of CD86+, MHC class II

+ cells comparable to treatment

with LPS (Figure3-2C).(Figure 3-2B). Infection with HK bacteria also increased the proportion

of positive cells albeit to a lesser degree than WT bacteria. Similar results were observed when

assessing the markers CD80+ and MHC class II

+ (Figure 3-2D). Infection with WT bacteria

increased the proportion of these cells compared to control cells. However, incubation with HK

bacteria did not increase the expression of these maturation markers above control cells (Figure

3-2E). Incubation with IL-6, which activated the STAT3 pathway comparable to H. pylori

infection, did not result in increased expression of maturation markers on DCs compared to

control cells. These studies demonstrate that H. pylori infection induces DC maturation.

Page 58: Ang Michelle 201006 MSc Thesis

FSC

103

102

101

100

0 200 400 600 800 1000

SSC

103

102

101

100

103102101100

CD

11b

A

49.7688.93

C

CD11c

B

103

102

101

100

CD86

MH

CII

Control LPS

103102101100

CD86

MH

CII

103

102

101

100

103102101100

CD86

MH

CII

IL-6

WT 7.13

103

102

101

100

103102101100

CD86

MH

CII

103

102

101

100

CD80

MH

CII

CD80

MH

CII

CD80

MH

CII

Control LPS IL-6

WT 7.13

CD80

103

102

101

100

MH

CII

103

102

101

100103102101100

103102101100103102101100 103102101100

103

102

101

100

103

102

101

100

103102101100

17.60 5.69

72.06 4.66

15.56 16.81

59.15 8.48

12.69 11.83

68.49 7.00

18.62 3.06

76.05 2.26

HK 7.13

103

102

101

100

103102101100

CD86

MH

CII

16.78 9.38

70.27 3.57

10.30 6.87

76.11 6.72

9.94 12.53

70.69 6.48

7.41 10.15

79.57 2.86

10.45 4.17

79.28 6.11

HK 7.13

103

102

101

100

CD80

MH

CII

103102101100

9.16 8.86

77.97 4.01

DE

*

*

45

Page 59: Ang Michelle 201006 MSc Thesis

46

Figure 3-2: H. pylori infection upregulates surface expression of DC maturation markers.

A) BMDC purity was determined using staining for CD11c+ and CD11b

+ and analyzed using

FACS analysis. B) DCs were incubated with either E. coli LPS (1μg/ml), IL-6 (100ng/ml), WT

bacteria (MOI 25:1) or HK bacteria for 24h. FACS analysis was used to determine the proportion

of cells positive for maturation markers MHC class II and CD86. Cells in upper right quadrant of

each scatter plot demonstrate the percentage of cells positive for both maturation markers. C)

Quantification of CD86+ and MHC class II

+ cells normalized to control. D) Scatter plots

represent cells stained with MHC class II and CD80. Upper right quadrants represent cells

positive for both surface markers. E) Quantification of CD80+ and MHC class II

+ cells. Columns,

mean; bars, SE;*, P < 0.05 using one way ANOVA, n=5.

Page 60: Ang Michelle 201006 MSc Thesis

47

H. pylori stimulated DCs induce an unusual cytokine profile

To determine the functional effects of H. pylori induced DC maturation, the cytokine expression

profile was assessed using the corresponding culture supernatants of BMDCs. Using a Luminex

multiplex panel, we examined a Th1/Th2 panel of cytokines. Of all the cytokines profiled in this

kit (IL-2, IL-4, IL-5, IL-10, IL-12, IL-17 and IFN-γ) only IL-12 (p40/p70) and IL-10 had levels

within detectable limits (Figure 3-3A). In comparison with supernatants obtained from either

control or IL-6 treated cells, WT infection increased the levels of both IL-12 and IL-10.

Supernatants from DCs incubated with HK bacteria showed a slight increase in IL-10 secretion

but not to the same levels as cells incubated with WT bacteria as results were not statistically

significant. When we compared the ratio of IL-10:IL-12 (Carrieri et al., 2008;Hwang et al.,

2007;Stax et al., 2008) infection with WT H. pylori increased the IL-10:IL-12. In contrast, the

proinflammatory E. coli LPS exerted an increased IL-12:IL-10 ratio (Figure 3-3B).

To further assess the cytokine profiles we employed ELISA-based assays. We first confirmed the

increase in IL-10 and IL-12 p40 with WT bacteria stimulation with ELISA analysis and an

increased immunosuppressive ratio of IL-10:IL-12 (Figure 3-4A, B). We could not detect an

increase in IL-12 p70 using the ELISA based assay. Additionally, we also saw an increase in IL-

6 and TNF-α stimulation with WT infection. (Figure 3-4A).

Page 61: Ang Michelle 201006 MSc Thesis

**

****

A

B

48

Page 62: Ang Michelle 201006 MSc Thesis

49

Figure 3-3: Luminex assay of H. pylori induced DC cytokine secretion.

A) Luminex milliplex assay was used to assess cytokine secretion for Th1/Th2 cytokines (IL-2,

IL-4, IL-5, IL-10, IL-12, IL-17 and IFN-γ in cell supernatants obtained from BMDCs incubated

with either E. coli LPS (1μg/ml), IL-6 (100ng/ml) WT or HK bacteria (MOI 25:1) for 24h.

Columns, means; bars, SE; *, P < 0.05; **, P < 0.01 using one-way ANOVA (n=3). B) Graphs

depict ratios of mean IL-10:IL-12 and IL-12:IL-10.

Page 63: Ang Michelle 201006 MSc Thesis

**

**

*

**AA

B

50

Page 64: Ang Michelle 201006 MSc Thesis

51

Figure 3-4: Determination of H. pylori induced DC cytokine secretion by ELISA based

assay.

A) ELISA was used to detect cytokine secretion for IL-10, IL-12, IL-6 and TNF-a in cell

supernatants obtained from BMDCs incubated with either E. coli LPS (1μg/ml), IL-6 (100ng/ml)

WT or HK bacteria (MOI 25:1) for 24h. Increases in IL-12p40, IL-10, TNF-α, and IL-6 were

detected with ELISA assays. Increases in IL-10 and TNF-α secretion were significant over

control. Columns, means; bars, SE; *, P < 0.05; **, P < 0.01 using one-way ANOVA (n=3). B)

Graph depicts ratios of IL-10:IL-12 and IL-12:IL-10.

Page 65: Ang Michelle 201006 MSc Thesis

52

H. pylori induced DC maturation and cytokine release is dependent on activation of the

STAT3 pathway.

To determine if the activation of the STAT3 pathway was responsible for the DC phenotype

observed, we employed the STAT3 inhibitor S3I-201. S3I-201 specifically inhibits the SH2

domains on STAT3 monomers, preventing dimerization and phosphorylation of the STAT3

pathway. S3I-201 inhibits constitutive Stat3 DNA-binding and transcriptional activities but has

no effect on Src activation or the EGFR-mediated activation of the Erk1/2(MAPK) pathway,

indicating that this inhibitor is specific for the STAT3 pathway (Zhang et al., 2010).

Cells were pretreated with S3I-201 for 30 min followed by incubation with WT bacteria or IL-6

treatment and the DC phenotype determined. Phosphorylation of STAT3 was inhibited in cells

pretreated with the STAT3 inhibitor and infected with WT H. pylori as assessed by

immunoblotting (Figure 3-5A). In addition, a decrease in expression of the DC maturation

markers was observed in comparison to cells infected with WT bacteria alone (Figure 3-5B,

n=2). Furthermore, S3I-201 pre-treatment prevented H. pylori-mediated secretion of IL-12p40,

IL-10 and TNF-α (Figure 3-5C). S3I-201 pre-treatment also resulted in a decrease in levels of

IL-6, although this did not reach significance. In addition, the IL-10:IL-12 ratio observed with

WT bacteria alone was reduced with S3I-201 treatment, (Figure 3-5D).

Page 66: Ang Michelle 201006 MSc Thesis

A

B

C

D

*** ***

** **

IL-6

WT 7

.13S3I-

201 +

WT 7

.13

PSTAT3

STAT3Con

trol

53

Page 67: Ang Michelle 201006 MSc Thesis

54

Figure 3-5: H. pylori induced DC maturation and cytokine secretion is dependent on

activation of the STAT3 pathway.

A) Western blot image of DC cell lysates obtained from cells pretreatment with S3I- 201 for

30min followed by incubation with WT bacteria for 24h. Corresponding blot below demonstrates

native STAT3 levels. B) Cells were incubated with WT bacteria (MOI 25:1) or S3I-201 (25μM)

(30 min pretreatment) plus WT bacteria for 24 h. Cells were assessed via flow cytometry for

expression of surface markers MHC class II, CD86 and CD80. Graphs represent cells positive

for either MHC class II and CD86 (left) or MHC class II and CD80 (right) (n=2). C) ELISA

analysis of DC culture supernatants. Columns, means; bars, SE; **, P < 0.01; ***, P < 0.001

using one-way ANOVA (n=3). D) Ratios of IL-10:IL-12 from mean ELISA

Page 68: Ang Michelle 201006 MSc Thesis

55

S3I-201 does not affect DC viability.

To ensure that treatment with S3I-201 does not affect DC viability, an MTT assay was utilized.

BMDCs were grown and infected for 24h as described. Viable cells contain mitochondrial

dehydrogenase, which cleaves the tetrazolium substrate ring, yielding purple formazan crystals.

Analysis demonstrated that cell viability is not affected with S3I-201 infection (Figure 3-6).

Page 69: Ang Michelle 201006 MSc Thesis

56

Page 70: Ang Michelle 201006 MSc Thesis

57

Figure 3-6: S3I-201 does not affect BMDC viability.

MTT analysis of DCs. DCs were incubated with IL-6 (100ng/ml), WT bacteria (MOI 25:1) or

S3I-201 (25μM) plus WT bacteria for 24 h. Spectrophotometric measurement of cell viability by

mitochodrial dehydrogenase activity which converts MTT to a water-insoluble colored formazan

derivative which is then solubilized in acidic isopropanol.

Page 71: Ang Michelle 201006 MSc Thesis

58

DISCUSSION

An increasing body of work is emerging studying the effects of H. pylori on DCs, linking

alterations in DC response to immune evasion. Despite the increasing interest, the exact

mechanisms by which H. pylori affects DC function is not known. In this study, we demonstrate

for the first time that H. pylori activates the STAT3 pathway in DCs. Additionally, we show that

H. pylori induces DC maturation as assessed by upregulation of the maturation markers MHC

class II, CD80 and CD86. Finally we show that H. pylori infected DCs cytokine profile shows an

enhanced IL-10: IL-12 ratio indicating a immunoregulatory phenotype. Both the DC maturation

and phenotype was dependent on STAT3 activation suggesting that STAT3 activation in DCs

may be a mechanism by which the bacterium subverts the host immune response.

We have shown that H. pylori mediated STAT3 activation in DCs occurs independently of the

CagA virulence factor and that live bacteria are required for maximal activation. However, our

studies also implicate a role for heat stable factors in this activation (e.g. LPS). In support of this

finding, Samavati et al. demonstrated that E. coli LPS can activate the STAT3 pathway in RAW

246.7 cells and BMDCs, leading to increased IL-1β, IL-6 and TNF-α production (Samavati et al.,

2009).

Use of the new inhibitor S3I-201, which blocks the phosphotyrosine binding region of the SH2

domain on STAT3, demonstrates the necessary involvement of STAT3 activation in H. pylori

induced DC maturation and cytokine release. Near-complete reversal of DC maturation and

cytokine release was seen when cells were pretreated with S3I-201 before WT incubation,

demonstrating for the first time the importance of STAT3 activation in these processes. Although

this inhibitor is thought to be specific it remains possible that other nonspecific effects may

occur. The use of additional mechanisms to inhibit STAT3, are now required to confirm the

STAT3 dependance on the observed DC phenotype.

STAT3 activation in immune cells is thought to have immunosuppressive effects. Lin and

collegues have demonstrated that Salmonella enteric serovar Typhimurium activates STAT3 in

macrophages, limiting excessive inflammatory responses and providing a hypothesized

mechanism of Salmonella’s immune evasion (Lin and Bost, 2004). Previous studies have

demonstrated that increased STAT3 activation in DCs can limit the inflammatory responses of

bacterial products. In liver DCs, STAT3 activation is upregulated. When challenged with

Page 72: Ang Michelle 201006 MSc Thesis

59

bacterial LPS, IL-6 release is noted, however due to elevated STAT3, DC activation is limited

(Lunz, III et al., 2007). STAT3 activation inhibits the production of DC IL-12, reducing

proinflammatory effects and downregulating the Th1 response (Wang et al., 2004). STAT3

inhibits IL-12p35 gene expression in tumour-associated DCs but the paralysis of tumour DCs

into an immature state has been shown to be reversed by blocking IL-10 activity, allowing de

novo IL-12 production (Vicari et al., 2002).

Additionally, STAT3 signalling has been linked to decreased tumour surveillance, decreasing the

ability of DCs to mature in a tumour environment (Lin et al., 2010c). STAT3 has also been

shown to inhibit DC maturation through the secretion of inhibitory IL-10, promoting the

development of inhibitory DCs (Corinti et al., 2001). Due to the role of STAT3 in directing DC

tumour immunosurveillance, our studies may demonstrate a mechanism by which H. pylori alters

DC signalling to induce tumourigenesis.

In our studies, we see a paradoxical increase in both IL -10 and IL-12 with H. pylori stimulated

STAT3 activation. However increases were only seen in IL-12p40, and not IL-12p70 (data not

shown) in our studies. IL-12p70 is the biologically active form of IL-12 composed of two

covalently linked chains of 40 and 35 kDa (IL-12p40 and IL-12p35) and is associated with

increased proinflammatory responses. Since in our studies, only IL-12p40 was induced, we

hypothesize that the increase in IL-12 with H. pylori infection does not induce a pro-

inflammatory response due to the lack of IL-12p70 formation, instead producing only IL-12p40.

Instead, IL-10 secretion is of greater importance. We report an increased IL-10:IL-12p40 ratio

observed with H. pylori infected DCs when compared to proinflammatory E.coli LPS. These

observations indicate a shift in importance from a Th1-IL-12 phenotype to an IL-10 response,

especially when noting the lack of IL-12p70. TNF-α elevation in response to H. pylori infection

seen in our studies may appear contradictory as TNF-α is known to be a proinflammatory

cytokine and associated with a Th1 response. However, evidence shows that TNF-α has also

been proven to be critical for the generation of IL-10 producing CD4+ T cells during the antigen

presentation by immature DCs, therefore providing an immunosuppressive role for TNF-α in our

DCs (Hirata et al., 2010).

Recently, activation of the STAT3 pathway has been linked to the development of a new class of

inflammation, the IL-23/Th17 response. The presence of only IL-12p40 can have increased

Page 73: Ang Michelle 201006 MSc Thesis

60

significance when considering that IL-23 shares the p40 chain and some properties with IL-12

and enhances Th17 responses. Reports have indicated that shifts from a anticarcinogenic IL-12

response to a pro-carcinogenic IL-23 is regulated through STAT3 signalling, leading to the

development of T-regulatory (Treg) cells, which would also induce IL-10 expression

(Kortylewski et al., 2009). We thus hypothesize that the increase in IL-10, IL-12p40 and IL-

10:IL-12 preferentially induces the development of Treg cells and possibly activation of the

Th17 pathway.

In conclusion, this study indentifies that H. pylori induces activation of the STAT3 signalling

pathway in DCs which is responsible for an induction of DC maturation, as well as an altered

cytokine profile, shifting the balance toward a potential Treg/Th17 polarization. These studies

indicate a novel mechanism by which H. pylori alters the innate immune response to prevent

elimination by the host and also potentially subvert tumour surveillance. Thus, the findings from

this study set the stage for investigating the effect of STAT3 inhibition on disease outcome

during H. pylori infection.

Page 74: Ang Michelle 201006 MSc Thesis

61

Chapter 4 Helicobacter pylori’s VacA Toxin Induces Reactive Oxygen Species

Production and Alters Parkin Distribution

INTRODUCTION

Oxidative stress has been noted in many tumours and is caused by an increase in the generation

of reactive oxygen species (ROS) alongside a decreased ability of the cell to clear these ROS.

High levels of ROS are associated with damage of proteins, lipids and DNA and all major

cellular constituents, whereas low ROS levels has been linked to the activation of intracellular

pathways leading to proliferation, promoting angiogenesis and metastasis (Droge,

2002;Weinberg and Chandel, 2009). ROS generation occurs with oxygen reduction, resulting in

the formation of the hydroxyl radical (•OH) and superoxide (•O2), which are easily integrated

into the activation of signalling pathways and react with iron to generate hydroxyl radicals

(Imlay et al., 1988).

Several sources of ROS have been identified. NADPH oxidase is one of the best studied sources

of •O2, catalyzing the production of •O2 from oxygen and NADPH (Lambeth, 2004). The

mitochondrial electron transport chain is also a major site of non-enzymatic •O2 formation, with

•O2 being created at complexes I, II and III. Additionally, the H2O2 created in these steps can be

further converted to •OH after contact with iron. Mitochondrial production of ROS is also an

important factor for the overall ROS level within a cell because it underlies oxidative damage in

many pathologic conditions, contributing to retrograde redox signalling from organelle to the

cytosol and the nucleus. Accumulation of mitochondrial derived ROS can lead to lipid

peroxidation, mitochondrial dysfunction, apoptosis and necrosis (Murphy, 2009). Additionally,

the ROS produced by the mitochondria have been reported to regulate autophagy and autophagic

cell death, as autophagy is induced by rotenone, H2O2, and superoxide (Chen and Gibson, 2008).

Recently, Parkin a gene first implicated in juvenile Parkinsonism has been shown to be recruited

to damaged, depolarized mitochondria selectively targeting these mitochondria for degradation

by autophagy (Narendra et al., 2008). Parkin is an E3-ligase, responsible for conjugating

ubiquitin to proteins, targeting them for degradation via the ubiquitin-proteasome degradation

pathway, where misfolded proteins are transported to aggresomes whose clearance is facilitated

by autophagy (Farrer, 2006;Olzmann and Chin, 2008). Mutations in the Parkin gene result in the

Page 75: Ang Michelle 201006 MSc Thesis

62

development of hepatocellular carcinoma, and the loss of Parkin contributes to the

overproliferation of hepatocytes (Ikeuchi et al., 2009). These combined observations indicate

that the Parkin gene may be important in tumourigenesis and cell cycle regulation in epithelial

cells (Farrer, 2006). Studies using Parkin knock-out mice demonstrated deficiencies in proteins

involved in mitochondrial function and oxidative stress, including reductions in proteins

involved in complexes I and IV (Palacino et al., 2004). Parkin deficient mice also exhibited

decreased levels of proteins involved in protection from oxidative stress, as well as decreased

serum antioxidant capacity and increased protein and lipid peroxidation, indicating that Parkin

mutations are associated with mitochondrial dysfunction (Palacino et al., 2004).

We have shown that H. pylori’s VacA activates the autophagy pathway. However the exact

mechanisms involved remain unknown. VacA localizes to mitochondria where it forms channels

in the membrane, reducing mitochondrial transmembrane potential, causing mitochondrial

dysfunction and cytochrome c release (Galmiche et al., 2000;Willhite et al., 2003;Willhite and

Blanke, 2004a). Therefore, we hypothesized that VacA may induce increased mitochondrial

derived ROS and Parkin redistribution to trigger autophagy. Therefore we determined if VacA

altered ROS production and Parkin distribution in gastric epithelial cells in vitro.

RESULTS

H. pylori’s VacA toxin induces ROS production

To determine if VacA increased ROS production, AGS cells were treated for 4 hours with

inactive or activated pure toxin and ROS measured using CM-H2DCFDA, a compound that

fluoresces when in contact with ROS, and assessed by FACS. As a positive control cells were

incubated with H2O2 (200µM, 40min). Activated VacA induced increased ROS production

comparable to levels seen in cells treated with H2O2 (Figure4 -1B). In marked contrast, ROS

levels were not increased in cells treated with inactive VacA toxin and resembled control

untreated cells (Figure 4-1A, B). We next determined if antioxidants could reduce VacA-

mediated ROS production. The antioxidants DPI, rotenone and α-tocopherol all failed to prevent

ROS induction (data not shown). However, in cells pre-treated with 5mM N-acetyl-cysteine

(NAC) VacA-mediated ROS was prevented (Figure 4-1A, B). Taken together, these results

demonstrate that VacA induces the production of ROS, which can be inhibited using NAC

treatment.

Page 76: Ang Michelle 201006 MSc Thesis

A

NAC

103102101100

100

60

20

80

0

40% o

f max

FL-1

12.2824.33

NAC + pure toxin

103102101100

100

60

20

80

0

40% o

f max

FL-1

12.2829.53

0 200 400 600 800 1000

FSC

SSC

0

200

400

600

800

1000

H2O2

% o

f max

FL-1

12.2874.81

103102101100

100

60

20

80

0

40

Pure VacA Toxin%

of m

ax

12.2859.25

FL-1103102101100

100

60

20

80

0

40

Inactive VacA Toxin

103102101100

100

60

20

80

0

40% o

f max

FL-1

12.2825.44

FL-1

# o

f cel

ls

Control

200

100

0

300

103102101100

12.28

B**

* *

63

Page 77: Ang Michelle 201006 MSc Thesis

64

Figure 4-1: VacA pure toxin induces ROS production in AGS cells.

AGS cells were incubated with activated VacA pure toxin or inactive toxin for 4h. Where

indicated, NAC (5mM) was utilized alone, or with pure toxin. Cells incubated with H2O2

(200μM, 30min) served as a positive ROS control. Cells A) ROS production was measured by

using a redox-sensitive dye (CM-H2DCFDA) on live cells, followed by flow cytometry analysis.

Overlaid curves indicate fluorescence curve for condition stated (green) versus control curve

(red) with corresponding numbers representing the percentage of ROS + cells assessed within the

gated region. B) Graph shows quantification of ROS experiments. Fold increase is expressed as

ratio of the %ROS+ cells in each condition/control. Columns, means; bars, SE; *, P < 0.05, **, P

< 0.01, using one-way ANOVA (n=3).

Page 78: Ang Michelle 201006 MSc Thesis

65

Inhibition of VacA induced ROS does not prevent VacA induced autophagy.

To determine if VacA induced ROS is responsible for VacA-mediated autophagy, we used

immunofluorescence to detect the redistribution of cytosolic LC3I to autophagic, membrane-

bound LC3II puncta. AGS cells were transfected with LC3-GFP plasmid and treated with EBSS

starvation conditions (a potent inducer of autophagy), or inactive or activated VacA pure toxin

for 4 hours in the presence or absence of NAC. As shown previously, starvation was sufficient to

induce the formation of autophagosomes indicated by green puncta formation (Figure 4-2A,

arrowheads). Similarly active but not inactive VacA induced the formation of LC3 puncta

(Figure 4-2A). However, treatment with NAC prior to incubation with active VacA did not

reduce the percent of autophagic cells (Figure 4-2A, B). These results suggest that inhibition of

VacA induced ROS does not alter VacA induced autophagy, indicating that ROS production and

autophagy induction by VacA occur independently.

Page 79: Ang Michelle 201006 MSc Thesis

Control Starvation

Pure VacA Toxin Inactive VacA Toxin

NAC NAC + Pure VacA

LC3-GFP

LC3-GFP

LC3-GFP

LC3-GFP

LC3-GFP

LC3-GFPContro

l

Starva

tion

Pure Toxin

Inactiv

e Toxin NAC

NAC + pure

toxin0

10

20

30

40

50

60

70

% A

utop

hagi

c ce

lls

*

*** ***

***

A

B

66

Page 80: Ang Michelle 201006 MSc Thesis

67

Figure 4-2: VacA induced ROS production is not responsible for VacA induced autophagy.

A) Confocal mircographs of AGS cells transfected with LC3-GFP (green) incubated with

activated VacA pure toxin, inactive toxin, NAC alone and NAC pretreatment followed by pure

toxin incubation. Starvation was used as a positive control for autophagy. Autophagy induction

is denoted by the formation of LC3 puncta (white arrowheads). Scale bars equals 15 μM. B)

Quantification of the percent autophagic cells for each treatment. Columns, means; bars, SE; *, P

< 0.05, ***, P < 0.001, using one-way ANOVA (n=4).

Page 81: Ang Michelle 201006 MSc Thesis

68

VacA induced autophagosomes do not colocalize with the mitochondria

VacA can localize to mitochondria and induce a variety of effects including increasing

mitochondrial permeability and the release of cytochrome c (Galmiche et al., 2000;Kimura et al.,

1999). Mitochondrial damage can induce mitochondrial specific autophagy (termed mitophagy),

particularly if the mitochondrial membrane potential is altered (Blanke, 2005;Kim et al., 2007b).

Therefore, due to VacA’s ability to alter the mitochondria, we next determined if VacA induced

mitophagy.

AGS cells transfected with LC3-GFP and the mitochondria marker Mito-RFP which fluoresces

in viable mitchondria were incubated with VacA pure toxin for 4h. Cells were then stained with

HSP60 (a mitochondrial marker staining the mitochondrial matrix) antibody with Cy5

conjugation to ensure all mitochondria were labelled. Immunofluorescence was used to

determine colocalization of the different markers.

Under starvation conditions, LC3-GFP puncta did not colocalize with mitochondrial markers as

seen in the merged image, indicating that the autophagosomes were not associated with the

mitochondria (Figure 4-3). Similarly, in cells treated with active purified VacA the majority of

autophagosomes did not colocalize with mitochondrial markers (Figure 4-3). Furthermore, NAC

pretreatment did not affect the degree of mitochondrial and LC3 colocalization (Figure 4-3).

Therefore, VacA induced autophagy does not selectively target mitochondria, to induce

mitophagy.

Page 82: Ang Michelle 201006 MSc Thesis

Control

Starvation

Pure VacA Toxin

Inactive VacA Toxin

NAC

NAC + Pure VacA Toxin

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

LC3-GFP Mito-RFP Hsp60-Cy5 LC3-GFPMito-RFPHsp60-Cy5

69

Page 83: Ang Michelle 201006 MSc Thesis

70

Figure 4-3: VacA induced LC3 aggregates do not colocalize with mitochondrial antibodies.

Confocal micrographs of AGS cells expressing LC3-GFP (green) and Mito-RFP (red) treated for

4h with either starvation media, pure activated VacA toxin, inactive toxin, NAC alone or NAC

pretreatment for 30min followed by incubation with pure toxin. HSP60 was immunolabelled

utilizing Cy5 (blue) conjugated secondary antibody. Arrows point to autophagosome formation.

Bars equal 15μM. Insets represent magnification of boxed area. Experiment was performed 3

times with representative images from one experiment shown.

Page 84: Ang Michelle 201006 MSc Thesis

71

H. pylori’s VacA toxin causes Parkin redistribution that is not dependent on VacA induced

autophagy

Although we did not demonstrate that VacA induced mitophagy, this process may be cell type

specific. Therefore we next determined if VacA mediated effects on mitochondria may alter

Parkin distribution. AGS cells were transfected with LC3-RFP and Parkin-YFP plasmids and

infected for 4h. Rotenone (50μM), which causes inhibition of the electron transport chain at

complex II and thus mitochondrial instability, was used as a positive control for Parkin

aggregation. Cells were assessed via spinning disc microscopy to determine if Parkin and LC3

aggregates colocalize. As expected, a diffuse cytosolic distribution of LC3 and Parkin was

detected in control cells (Figure 4-4A). Rotenone induced Parkin redistribution but no change in

LC3 compared to control cells (Figure 4-4A). Cells treated with inactive toxin resembled control

cells, (Figure 4-4A). In contrast, in cells incubated with pure toxin LC3 and Parkin were

redistributed in aggregates(Figure 4-4A, B). However significant colocalization of LC3 and

Parkin aggregates was not detected. Furthermore, LC3 and Parkin aggregation was not always

seen in tandem, as some cells contain only LC3 aggregation, while others have only Parkin

redistribution (Figure 4-4C). In addition, NAC treatment which reduced ROS failed to prevent

pure toxin induced Parkin redistribution. These results indicate that VacA treatment can cause

Parkin redistribution. However VacA’s ability to induce autophagy appears to be independent of

Parkin aggregation and VacA induced ROS.

Page 85: Ang Michelle 201006 MSc Thesis

Control

Starvation

NAC

NAC + Pt

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

Inactive Toxin

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

Pure Toxin

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

Contro

l

Starva

tion

Roteno

ne

Pure V

acA Tox

in

Inacti

ve Tox

inNAC

NAC + Pure

Toxin

05

101520253035404550556065

% Autophagic% (+)Parkin aggregation

% o

f cou

nted

cel

ls

AB

Control

Starva

tion

Rotenone

Pure Toxin

Inactiv

e Toxin

0

10

20

30

40

% c

ells

with

par

kin

aggr

egat

ion

C

**

Rotenone

LC3-RFP Parkin-YFP LC3-RFPParkin-YFP

72

Page 86: Ang Michelle 201006 MSc Thesis

73

Figure 4-4: VacA pure toxin induces redistribution of Parkin that is independent of its

induction of autophagy.

A) Confocal micrographs of AGS cells expressing Parkin-YFP (green) and LC3-RFP (red)

treated with pure activated VacA toxin, inactive toxin, NAC alone or NAC pretreatment for

30min followed by incubation with pure toxin for 4h. Arrows represent autophagosome

formation in LC3-RFP panels or Parkin aggregates in Parkin-YFP panels. Insets show

magnifications of Parkin redistribution and autophagy formation. B) Graph shows the percent of

cells with Parkin aggregation. Columns, means; bars, SE; **, P < 0.01, using one-way ANOVA

(n=4). C) Graph demonstrates percent of autophagic cells (light grey) or Parkin redistributed

cells (dark grey). Columns, means; bars, SE (n=4).

Page 87: Ang Michelle 201006 MSc Thesis

74

VacA induced Parkin aggregation does not colocalize with the mitochondria.

Since Parkin aggregation has been shown to selectively target the mitochondria for autophagic

degradation, we next examined if VacA toxin induces Parkin recruitment to the mitochondria.

AGS cells were transfected with LC3-RFP and Parkin-YFP plasmids and treated for 4h with

VacA pure toxin or starvation conditions (Figure 4-5). Cells were subsequently stained with

HSP60 Cy5 and examined using spinning disc microscopy. Colocalization between Parkin

aggregates and mitochondrial antibodies was not detected. Therefore, despite Parkin’s ability to

induce mitophagy, VacA induced Parkin aggregation does not lead to mitochondrial autophagy.

Page 88: Ang Michelle 201006 MSc Thesis

LC3-RFP Parkin-YFP HSP 60 Cy5

LC3-RFP Parkin-YFP HSP 60 Cy5

LC3-RFP Parkin-YFP HSP 60 Cy5

Control

Starvation

Pure Toxin

75

Page 89: Ang Michelle 201006 MSc Thesis

76

Figure 4-5: VacA induced Parkin aggregation does not target to the mitochondria.

Confocal micrographs of AGS cells expressing Parkin-YFP (green) and LC3-RFP (red) treated

for 4h with either starvation conditions or pure activated VacA toxin. Cells were stained for

HSP60 conjugated to Cy5 and colocalization between HSP60 and Park-YFP was not noted.

Insets show magnifications of Parkin redistribution and autophagy formation.

Page 90: Ang Michelle 201006 MSc Thesis

77

DISCUSSION

Autophagy is a mechanism used by host cells to eliminate intracellular pathogens, having a role

in antigen processing and influencing the host immune response. We have shown previously that

H. pylori’s VacA toxin is capable of inducing autophagy in gastric epithelial cells. VacA can

affect mitochondria. The toxin localizes to the mitochondria where it forms channels in its

membrane, disrupting mitochondrial membrane potential and inducing the release of cytochrome

c. Since alterations in mitochondria can induce autophagy we determined if VacA’s

mitochondrial effects induce the production of ROS, thereby inducing VacA induced autophagy.

In our studies, autophagosomes induced by VacA toxin did not colocalize with mitochondria

suggesting that VacA induced autophagy was not specifically targeting the mitochondria for

degradation. However, due to VacA’s ability to interact with the mitochondria, we further

investigated the role of ROS.

The production of ROS has been previously linked with H. pylori infection. Increased reactive

oxygen metabolites have been found in H. pylori positive patients, but not in H. pylori negative

patients (Davies et al., 1994). Additionally, H. pylori induces enhanced production of ROS in

gastric cells, and enhances membrane damage (Bagchi et al., 1996). H. pylori can also induce the

recruitment and activation of monocytes and neutrophils that create ROS (Mashimo et al., 2006).

H. pylori induces ROS with increasing levels of DCF fluorescence as well showing increasing

levels of MitoSOX Red fluorescence, an indicator of mitochondrial superoxide production

(Calvino-Fernandez et al., 2008). As well, an increase in ROS-mediated DNA oxidation is seen

in the gastric mucosa upon H. pylori infection, and these levels are reduced upon H. pylori

eradication (Katsurahara et al., 2009). Therefore, ROS production in association with H. pylori

infection is well established.

Our studies are the first to demonstrate that VacA alone can induce ROS production. The origin

of these ROS remains unknown. Lack of ROS inhibition by DPI, which specifically blocks

NADPH oxidase, indicates that the NOX-2 pathway is not responsible for ROS production in

VacA treatment despite being implicated in other antibacterial autophagic processes (Huang and

Brumell, 2009). We speculate that VacA interaction with mitochondria may be the source of

ROS induction. However, further studies are required to define the exact source of ROS.

Page 91: Ang Michelle 201006 MSc Thesis

78

ROS production can induce the autophagic process, with ROS being specifically required to

regulate Atg4 activity (Scherz-Shouval et al., 2007), and superoxide being responsible for

regulating autophagy (Chen et al., 2009). However the ROS production in our studies does not

seem to play a role in VacA induction of autophagy, as NAC (a whole-cell antioxidant)

treatment, which reduces ROS did not inhibit VacA induced autophagy. Host cells may induce

ROS to limit bacterial replication (Mastroeni et al., 2000). VacA increases intracellular bacterial

survival, (Terebiznik et al., 2006) therefore host cells may induce ROS production to limit VacA

induced intracellular survival and reduce microbial burden.

Since ROS production did not appear to be the cause of VacA induced autophagy, we next

looked at the E3-ligase Parkin to examine if VacA can induce Parkin aggregation. Parkin has

recently been linked to autophagy in a mitochondrial specific manner (Narendra et al., 2008).

Our studies demonstrate that VacA can cause re-distribution of Parkin. These Parkin aggregates

did not appear to colocalize with VacA (data not shown) or mitochondrial antibodies. The

significance of VacA induced Parkin aggregation is not yet known.

In summary, we demonstrate that VacA toxin induces ROS production in AGS cells. However

these ROS do not mediate autophagy. Parkin aggregation is also seen with VacA incubation, and

the implications of this are not yet known. Although the specific mechanisms by which H. pylori

induces autophagy are still unknown, knowledge that VacA induces ROS could potentially

provide an avenue by which host cells limit VacA bacterial survival. These studies provide the

platform for future studies that may elucidate the specific mechanisms by which autophagy is

induced, providing more insight into H. pylori’s effects on host immune response.

Page 92: Ang Michelle 201006 MSc Thesis

79

Chapter 5 Discussion and Future Directions

Affecting or altering the host immune response is crucial in the long term infection and survival

of H. pylori. Alterations in innate immune signalling ultimately determine the outcome of the

bacterial infection, orchestrating the response of secondary T-cells, and coordinating the adaptive

immune response.

Two key components of the innate immune response are the activation of DCs and the induction

of autophagy. Both play crucial roles in detecting and responding to pathogenic bacteria

infection and the manipulation of these responses by bacteria are crucial in their survival. In our

studies, we determined that H. pylori alters the responses of both the autophagic pathway and the

activation of DCs, providing possible mechanisms for immune subversion by the bacterium.

In DCs, H. pylori activates STAT3 and induces maturation and activation of a unique phenotype

of DCs, differing from that seen with IL-6 treatment or induced by E. coli LPS. The cytokine

profiles induced by H. pylori are skewed to IL-10 induced immunoregulatory responses, likely

preventing robust pro-inflammatory responses and allowing H. pylori to avoid elimination by the

host innate immune response. STAT3 regulates the activity of IL-10, as STAT3 knock-out DCs

are resistant to IL-10 suppression of DC activation, allowing enhanced inflammatory cytokine

secretion (Melillo et al., 2010b). Thus the increased IL-10 and STAT3 signalling we detected

may be crucial to the regulation of DCs, These developments may assist in producing STAT3

based therapies to prevent H. pylori long term colonization.

Our studies also indicate the potential involvement of enhanced IL-23 secretion in H. pylori

infected DCs as discussed in chapter 3. STAT3 signalling has been linked to the development of

a Th17 response, inducing IL-17 and IL-23 expression (Harris et al., 2007;Yang et al., 2007). IL-

23 has been implicated in other chronic disorders such as inflammatory bowel disease (IBD),

Crohn’s disease and colitis as IL-23 deficiency or blockade protects from Crohn’s disease as well

as human IBD (Abraham and Cho, 2009). Similarly, the proposed initiation of an IL-23/Treg

response may be a mechanism by which H. pylori promotes long term and chronic infection.

Furthermore, it has been shown that STAT3 signalling within the tumour microenvironment

induces IL-23, which was shown to be a procarcinogenic cytokine, while inhibiting

Page 93: Ang Michelle 201006 MSc Thesis

80

anticarcinogenic, IL-12, thereby shifting the balance of tumour immunity toward

carcinogenesis(Kortylewski et al., 2009). IL-23 also has been linked to tumourigenesis and the

development of cancer. It has also been demonstrated that IL-23 has an important role in the

production of IL-17 producing CD8+ T-cells leading to the development of tumourigenesis and

disease (Ciric et al., 2009). As well, antibody-mediated blockade of the receptor for IL-23

inhibits Bacteroides fragilis, a commensal colonic bacterium, STAT3 induced colitis, colonic

hyperplasia and tumour formation (Wu et al., 2009).

The STAT3 pathway has been linked to oncogenesis, as STAT3 is persistently activated in many

human cancers and transformed cell lines (Bromberg et al., 1999). Indeed, STAT3 expression

correlates with a poor prognosis in gastric cancer (Kim et al., 2009). H. pylori’s activation of

STAT3 in DCs could promote oncogenesis. Recent evidence indicates that dendritic cells can

integrate signals from the tumour microenvironment to modulate tumour immunity (Lin et al.,

2010b). STAT3 hyperactivation has been observed in DCs infiltrating tumours and decreases

their immunostimulatory capacity thereby facilitating tumour immune evasion (Cheng et al.,

2003). Indeed, specific targeted deletion of STAT3 in DCs in mice confirms the role for STAT3

as a negative regulator of DC function (Melillo et al., 2010a). Hyperactivation of STAT3 in DCs

increases IL-10 production, common in the tumour microenvironment and similar to the results

we observed in H. pylori treated DCs (Lin et al., 2010a). Our findings suggest that in addition to

epithelial STAT3 activation, STAT3 activation in DCs could also be involved in promotion of

carcinogenesis during chronic H. pylori infection.

Using a specific STAT3 inhibitor, S3I-201 we observed a reversal of STAT3 induced DC effects

in vitro. Previous studies demonstrate that inhibition of STAT3 with JSI-124 can inhibit the

growth of STAT3 hyperactivated cell lines in mice as well as reduce the tumour burden of

melanoma cells (Blaskovich et al., 2003). Therefore our studies set the stage for investigating the

role of STAT3 as a promoter of a pro-tumour microenvironment due to DC alteration in vivo.

Autophagy induction can function as an innate defence mechanism by killing intracellular

microorganisms attempting to establish a replicative niche in the host cytoplasm (Levine and

Deretic, 2007). However, previous studies have shown several pathogens such as Listeria

monocytogenes and Shigella flexneri are able to evade autophagy to promote intracellular

survival (Hussey et al., 2009;Orvedahl and Levine, 2009). Results from our lab suggest that

Page 94: Ang Michelle 201006 MSc Thesis

81

during acute exposure, VacA-induced autophagy reduces the effects of the toxin in host cells.

The exact mechanisms by which VacA induces autophagy remain unknown and were the focus

of chapter 4. We did not identify VacA-induced mitophagy but did determine that VacA could

also cause redistribution of Parkin, a protein which localizes to damaged mitochondria. Our

studies also demonstrated that VacA induces ROS production, however VacA induced

autophagy is not dependent on ROS production. The significance of VacA induced ROS is not

yet known. Increased ROS levels are known to cause oxidative DNA damage, potentially leading

to deregulation of tumour suppressing elements such as p53, and enhancing tumourigenesis.

Thus VacA induced ROS may contribute to H. pylori induced carcinogenesis.

Manipulations of the innate immune response have been implicated in shifting the balance to a

protumour type of immunity (Diacovich and Gorvel, 2010). Although H. pylori evades

elimination by the host immune response, chronic inflammation is associated with infection.

Chronic inflammation has been associated with increased tumour risk, is involved in polarizing

immunity toward those effectors that facilitate tumour growth (Ostrand-Rosenberg, 2008). Thus

alteration and manipulation of the innate immune response by H. pylori can not only result in

successful immune evasion thereby promoting chronic infection and also promoting

tumourigenesis.

Overall, our results demonstrate new and novel mechanisms by which H. pylori may alter the

host immune response to promote long term survival. By unravelling the various mechanisms by

which H. pylori alters the innate immune response there will be a greater understanding into its

colonization, chronic infection and disease outcomes, leading to the development of new

preventative and treatment strategies .

FUTURE DIRECTIONS

The results of our studies investigating the effects of H. pylori infection on DCs have uncovered

several future directions that need to be addressed. To further define the mechanism of STAT3

mediated DC activation, a more specific STAT3 inhibitor, SFI-001, has been developed, which

binds the SH2 domains on STAT3 monomers. This new inhibitor can more definitively

demonstrate if STAT3 is responsible for the DC phenotype we observe.

Page 95: Ang Michelle 201006 MSc Thesis

82

Due to the postulated involvement of IL-23, it would also be of interest to examine DC secretion

for presence of IL-23. At the time of our studies, a mouse IL-23 kit was not available to us,

however examination of this cytokine would be a crucial next step.

Another future consideration would look into the functional effect of H. pylori infected DCs on

T-cells to definitively determine if these DCs would lead to the development of T-regulatory

cells. H. pylori stimulated DCs could be incubated with T-cells to determine the effects on T-cell

proliferation, cytokine secretion, and finally determining the overall T-cell polarization. It would

also be of interest to determine if the DC cytokine secretion we observed would be sufficient to

polarize T-cells to Treg cells. T-cells could be stimulated with DC supernatant secretion and

assessed for functional changes.

Mellilo and colleagues have recently developed a DC targeted STAT3 conditional knock-out

(CKO) mouse to determine specific effects of STAT3 in DCs (Melillo et al., 2010c). Using these

DCs, we could further define the role of STAT3 activation by H. pylori. To extend these studies

to human subjects STAT3 activation in DCs could be confirmed via immunofluorescence in

tissue sections of H. pylori infected stomachs.

The purpose and importance of VacA induced ROS needs additional study as well. The role of

ROS as a mechanism to limit intracellular bacterial survival could be assessed, using intracellular

survival assays to determine if inhibiting ROS leads to increased bacterial survival. Other aspects

of VacA induced ROS should also be examined, as the mechanism of ROS production is not yet

known. The lack of ROS inhibition by DPI, suggests that ROS production is not dependant on

the NOX/NADPH pathway. Use of other antioxidants could be used to characterize the ROS, as

well as examination via RTPCR for upregulation of genes related to oxidative stress.

The effects of VacA on endogenous Parkin needs further study. Little is known about the

potential effects of Parkin in epithelial cells as most studies focus on neuronal cells.

Additionally, although we did not see colocalization of VacA with Parkin at 4h, later timepoints

should be assessed to confirm these findings. Additionally, to determine the relevance of Parkin

aggregation siRNA to inhibit Parkin or essential autophagy genes would allow us to determine

definitively if Parkin redistribution is linked to the autophagy pathway, or if autophagy is

essential for Parkin redistribution. Finally, it would also be important to confirm these findings in

tissue sections to determine if Parkin aggregation plays a role in H. pylori infected tissues.

Page 96: Ang Michelle 201006 MSc Thesis

83

References

References

Abraham, C. and Cho, J. H. (2009) IL-23 and autoimmunity: new insights into the pathogenesis

of inflammatory bowel disease. Annu Rev Med 60: 97-110.

Amieva, M. R. and El Omar, E. M. (2008) Host-bacterial interactions in Helicobacter pylori

infection. Gastroenterology 134: 306-323.

Amieva, M. R., Vogelmann, R., Covacci, A., Tompkins, L. S., Nelson, W. J., and Falkow, S. (5-

30-2003) Disruption of the epithelial apical-junctional complex by Helicobacter pylori CagA.

Science 300: 1430-1434.

Backhed, F., Rokbi, B., Torstensson, E., Zhao, Y., Nilsson, C., Seguin, D., Normark, S., Buchan,

A. M., and Richter-Dahlfors, A. (3-1-2003) Gastric mucosal recognition of Helicobacter pylori is

independent of Toll-like receptor 4. J Infect Dis 187: 829-836.

Bagchi, D., Bhattacharya, G., and Stohs, S. J. (1996) Production of reactive oxygen species by

gastric cells in association with Helicobacter pylori. Free Radic Res 24: 439-450.

Banchereau, J. and Steinman, R. M. (3-19-1998) Dendritic cells and the control of immunity.

Nature 392: 245-252.

Blanke, S. R. (2005) Micro-managing the executioner: pathogen targeting of mitochondria.

Trends Microbiol 13: 64-71.

Blaskovich, M. A., Sun, J., Cantor, A., Turkson, J., Jove, R., and Sebti, S. M. (3-15-2003)

Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of

transcription 3 signaling pathway inhibitor with potent antitumor activity against human and

murine cancer cells in mice. Cancer Res 63: 1270-1279.

Boncristiano, M., Paccani, S. R., Barone, S., Ulivieri, C., Patrussi, L., Ilver, D., Amedei, A.,

D'Elios, M. M., Telford, J. L., and Baldari, C. T. (12-15-2003) The Helicobacter pylori

vacuolating toxin inhibits T cell activation by two independent mechanisms. J Exp Med 198:

1887-1897.

Bourzac, K. M. and Guillemin, K. (2005) Helicobacter pylori-host cell interactions mediated by

type IV secretion. Cell Microbiol 7: 911-919.

Bromberg, J. F., Wrzeszczynska, M. H., Devgan, G., Zhao, Y., Pestell, R. G., Albanese, C., and

Darnell, J. E., Jr. (8-6-1999) Stat3 as an oncogene. Cell 98: 295-303.

Bronte-Tinkew, D. M., Terebiznik, M., Franco, A., Ang, M., Ahn, D., Mimuro, H., Sasakawa,

C., Ropeleski, M. J., Peek, R. M., Jr., and Jones, N. L. (1-15-2009) Helicobacter pylori

Page 97: Ang Michelle 201006 MSc Thesis

84

cytotoxin-associated gene A activates the signal transducer and activator of transcription 3

pathway in vitro and in vivo. Cancer Res 69: 632-639.

Calvino-Fernandez, M., Benito-Martinez, S., and Parra-Cid, T. (2008) Oxidative stress by

Helicobacter pylori causes apoptosis through mitochondrial pathway in gastric epithelial cells.

Apoptosis 13: 1267-1280.

Carrieri, P. B., Ladogana, P., Di Spigna, G., de Leva, M. F., Petracca, M., Montella, S.,

Buonavolonta, L., Florio, C., and Postiglione, L. (2008) Interleukin-10 and interleukin-12

modulation in patients with relapsing-remitting multiple sclerosis on therapy with interferon-beta

1a: differences in responders and non responders. Immunopharmacol Immunotoxicol 30: 1-9.

Chen, Y., Azad, M. B., and Gibson, S. B. (5-1-2009) Superoxide is the major reactive oxygen

species regulating autophagy. Cell Death Differ.

Chen, Y. and Gibson, S. B. (2-16-2008) Is mitochondrial generation of reactive oxygen species a

trigger for autophagy? Autophagy 4: 246-248.

Cheng, F., Wang, H. W., Cuenca, A., Huang, M., Ghansah, T., Brayer, J., Kerr, W. G., Takeda,

K., Akira, S., Schoenberger, S. P., Yu, H., Jove, R., and Sotomayor, E. M. (2003) A critical role

for Stat3 signaling in immune tolerance. Immunity 19: 425-436.

Churin, Y., Al Ghoul, L., Kepp, O., Meyer, T. F., Birchmeier, W., and Naumann, M. (4-28-

2003) Helicobacter pylori CagA protein targets the c-Met receptor and enhances the motogenic

response. J Cell Biol 161: 249-255.

Ciric, B., El behi, M., Cabrera, R., Zhang, G. X., and Rostami, A. (5-1-2009) IL-23 drives

pathogenic IL-17-producing CD8+ T cells. J Immunol 182: 5296-5305.

Corinti, S., Albanesi, C., la Sala, A., Pastore, S., and Girolomoni, G. (4-1-2001) Regulatory

activity of autocrine IL-10 on dendritic cell functions. J Immunol 166: 4312-4318.

Correa, P. (4-2-2003) Bacterial infections as a cause of cancer. J Natl Cancer Inst 95: E3.

Correa, P. (2004) The biological model of gastric carcinogenesis. IARC Sci Publ 301-310.

Correa, P. and Houghton, J. (2007) Carcinogenesis of Helicobacter pylori. Gastroenterology

133: 659-672.

Cover, T. L. and Blanke, S. R. (2005) Helicobacter pylori VacA, a paradigm for toxin

multifunctionality. Nat Rev Microbiol 3: 320-332.

Cover, T. L. and Blaser, M. J. (2009) Helicobacter pylori in health and disease. Gastroenterology

136: 1863-1873.

Cunningham, M. D., Seachord, C., Ratcliffe, K., Bainbridge, B., Aruffo, A., and Darveau, R. P.

(1996) Helicobacter pylori and Porphyromonas gingivalis lipopolysaccharides are poorly

transferred to recombinant soluble CD14. Infect Immun 64: 3601-3608.

Page 98: Ang Michelle 201006 MSc Thesis

85

Davies, G. R., Simmonds, N. J., Stevens, T. R., Sheaff, M. T., Banatvala, N., Laurenson, I. F.,

Blake, D. R., and Rampton, D. S. (1994) Helicobacter pylori stimulates antral mucosal reactive

oxygen metabolite production in vivo. Gut 35: 179-185.

Dhar, S. K., Soni, R. K., Das, B. K., and Mukhopadhyay, G. (2003) Molecular mechanism of

action of major Helicobacter pylori virulence factors. Mol Cell Biochem 253: 207-215.

Diacovich, L. and Gorvel, J. P. (2010) Bacterial manipulation of innate immunity to promote

infection. Nat Rev Microbiol 8: 117-128.

Droge, W. (2002) Free radicals in the physiological control of cell function. Physiol Rev 82: 47-

95.

Farrer, M. J. (2006) Genetics of Parkinson disease: paradigm shifts and future prospects. Nat Rev

Genet 7: 306-318.

Ferrero, R. L. (2005) Innate immune recognition of the extracellular mucosal pathogen,

Helicobacter pylori. Mol Immunol 42: 879-885.

Galmiche, A., Rassow, J., Doye, A., Cagnol, S., Chambard, J. C., Contamin, S., de, Thillot, V,

Just, I., Ricci, V., Solcia, E., Van Obberghen, E., and Boquet, P. (12-1-2000) The N-terminal 34

kDa fragment of Helicobacter pylori vacuolating cytotoxin targets mitochondria and induces

cytochrome c release. EMBO J 19: 6361-6370.

Gauthier, N. C., Monzo, P., Gonzalez, T., Doye, A., Oldani, A., Gounon, P., Ricci, V., Cormont,

M., and Boquet, P. (4-23-2007) Early endosomes associated with dynamic F-actin structures are

required for late trafficking of H. pylori VacA toxin. J Cell Biol 177: 343-354.

Gebert, B., Fischer, W., Weiss, E., Hoffmann, R., and Haas, R. (8-22-2003) Helicobacter pylori

vacuolating cytotoxin inhibits T lymphocyte activation. Science 301: 1099-1102.

Gobert, A. P., Bambou, J. C., Werts, C., Balloy, V., Chignard, M., Moran, A. P., and Ferrero, R.

L. (1-2-2004) Helicobacter pylori heat shock protein 60 mediates interleukin-6 production by

macrophages via a toll-like receptor (TLR)-2-, TLR-4-, and myeloid differentiation factor 88-

independent mechanism. J Biol Chem 279: 245-250.

Goodwin, C. S., Armstrong, J. A., and Marshall, B. J. (1986) Campylobacter pyloridis, gastritis,

and peptic ulceration. J Clin Pathol 39: 353-365.

Greenlund, A. C., Morales, M. O., Viviano, B. L., Yan, H., Krolewski, J., and Schreiber, R. D.

(1995) Stat recruitment by tyrosine-phosphorylated cytokine receptors: an ordered reversible

affinity-driven process. Immunity 2: 677-687.

Guiney, D. G., Hasegawa, P., and Cole, S. P. (2003) Helicobacter pylori preferentially induces

interleukin 12 (IL-12) rather than IL-6 or IL-10 in human dendritic cells. Infect Immun 71: 4163-

4166.

Harris, T. J., Grosso, J. F., Yen, H. R., Xin, H., Kortylewski, M., Albesiano, E., Hipkiss, E. L.,

Getnet, D., Goldberg, M. V., Maris, C. H., Housseau, F., Yu, H., Pardoll, D. M., and Drake, C.

Page 99: Ang Michelle 201006 MSc Thesis

86

G. (10-1-2007) Cutting edge: An in vivo requirement for STAT3 signaling in TH17 development

and TH17-dependent autoimmunity. J Immunol 179: 4313-4317.

Hatakeyama, M. (2004) Oncogenic mechanisms of the Helicobacter pylori CagA protein. Nat

Rev Cancer 4: 688-694.

Hatakeyama, M. (9-15-2006) Helicobacter pylori CagA -- a bacterial intruder conspiring gastric

carcinogenesis. Int J Cancer 119: 1217-1223.

Hirano, T., Nakajima, K., and Hibi, M. (1997) Signaling mechanisms through gp130: a model of

the cytokine system. Cytokine Growth Factor Rev 8: 241-252.

Hirata, N., Yanagawa, Y., Satoh, M., Ogura, H., Ebihara, T., Noguchi, M., Matsumoto, M.,

Togashi, H., Seya, T., Onoe, K., and Iwabuchi, K. (2010) Dendritic cell-derived TNF-alpha is

responsible for development of IL-10-producing CD4+ T cells. Cell Immunol 261: 37-41.

Horvath, C. M. and Darnell, J. E. (1997) The state of the STATs: recent developments in the

study of signal transduction to the nucleus. Curr Opin Cell Biol 9: 233-239.

Huang, J. and Brumell, J. H. (2009) NADPH oxidases contribute to autophagy regulation.

Autophagy 5: 887-889.

Huang, J. and Klionsky, D. J. (8-1-2007) Autophagy and human disease. Cell Cycle 6: 1837-

1849.

Hussey, S., Travassos, L. H., and Jones, N. L. (2009) Autophagy as an emerging dimension to

adaptive and innate immunity. Semin Immunol 21: 233-241.

Hwang, S. A., Wilk, K. M., Bangale, Y. A., Kruzel, M. L., and Actor, J. K. (2007) Lactoferrin

modulation of IL-12 and IL-10 response from activated murine leukocytes. Med Microbiol

Immunol 196: 171-180.

Ikeuchi, K., Marusawa, H., Fujiwara, M., Matsumoto, Y., Endo, Y., Watanabe, T., Iwai, A.,

Sakai, Y., Takahashi, R., and Chiba, T. (11-1-2009) Attenuation of proteolysis-mediated cyclin E

regulation by alternatively spliced Parkin in human colorectal cancers. Int J Cancer 125: 2029-

2035.

Imlay, J. A., Chin, S. M., and Linn, S. (4-29-1988) Toxic DNA damage by hydrogen peroxide

through the Fenton reaction in vivo and in vitro. Science 240: 640-642.

Jung, H. S., Chung, K. W., Won, Kim J., Kim, J., Komatsu, M., Tanaka, K., Nguyen, Y. H.,

Kang, T. M., Yoon, K. H., Kim, J. W., Jeong, Y. T., Han, M. S., Lee, M. K., Kim, K. W., Shin,

J., and Lee, M. S. (2008) Loss of autophagy diminishes pancreatic beta cell mass and function

with resultant hyperglycemia. Cell Metab 8: 318-324.

Kao, J. Y., Rathinavelu, S., Eaton, K. A., Bai, L., Zavros, Y., Takami, M., Pierzchala, A., and

Merchant, J. L. (2006) Helicobacter pylori-secreted factors inhibit dendritic cell IL-12 secretion:

a mechanism of ineffective host defense. Am J Physiol Gastrointest Liver Physiol 291: G73-

G81.

Page 100: Ang Michelle 201006 MSc Thesis

87

Kao, J. Y., Zhang, M., Miller, M. J., Mills, J. C., Wang, B., Liu, M., Eaton, K. A., Zou, W.,

Berndt, B. E., Cole, T. S., Takeuchi, T., Owyang, S. Y., and Luther, J. (11-18-2009) Helicobacter

pylori Immune Escape Is Mediated by Dendritic Cell-Induced Treg Skewing and Th17

Suppression in Mice. Gastroenterology.

Katsurahara, M., Kobayashi, Y., Iwasa, M., Ma, N., Inoue, H., Fujita, N., Tanaka, K., Horiki, N.,

Gabazza, E. C., and Takei, Y. (2009) Reactive nitrogen species mediate DNA damage in

Helicobacter pylori-infected gastric mucosa. Helicobacter 14: 552-558.

Kido, M., Tanaka, J., Aoki, N., Iwamoto, S., Nishiura, H., Chiba, T., and Watanabe, N. (2010)

Helicobacter pylori promotes the production of thymic stromal lymphopoietin by gastric

epithelial cells and induces dendritic cell-mediated inflammatory Th2 responses. Infect Immun

78: 108-114.

Kim, D. J., Chan, K. S., Sano, S., and Digiovanni, J. (2007a) Signal transducer and activator of

transcription 3 (Stat3) in epithelial carcinogenesis. Mol Carcinog 46: 725-731.

Kim, D. Y., Cha, S. T., Ahn, D. H., Kang, H. Y., Kwon, C. I., Ko, K. H., Hwang, S. G., Park, P.

W., Rim, K. S., and Hong, S. P. (2009) STAT3 expression in gastric cancer indicates a poor

prognosis. J Gastroenterol Hepatol 24: 646-651.

Kim, I., Rodriguez-Enriquez, S., and Lemasters, J. J. (6-15-2007b) Selective degradation of

mitochondria by mitophagy. Arch Biochem Biophys 462: 245-253.

Kimura, M., Goto, S., Wada, A., Yahiro, K., Niidome, T., Hatakeyama, T., Aoyagi, H.,

Hirayama, T., and Kondo, T. (1999) Vacuolating cytotoxin purified from Helicobacter pylori

causes mitochondrial damage in human gastric cells. Microb Pathog 26: 45-52.

Kisseleva, T., Bhattacharya, S., Braunstein, J., and Schindler, C. W. (2-20-2002) Signaling

through the JAK/STAT pathway, recent advances and future challenges. Gene 285: 1-24.

Klionsky, D. J. and Emr, S. D. (12-1-2000) Autophagy as a regulated pathway of cellular

degradation. Science 290: 1717-1721.

Kortylewski, M., Xin, H., Kujawski, M., Lee, H., Liu, Y., Harris, T., Drake, C., Pardoll, D., and

Yu, H. (2-3-2009) Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor

microenvironment. Cancer Cell 15: 114-123.

Kortylewski, M. and Yu, H. (2007) Stat3 as a potential target for cancer immunotherapy. J

Immunother (1997 ) 30: 131-139.

Kranzer, K., Eckhardt, A., Aigner, M., Knoll, G., Deml, L., Speth, C., Lehn, N., Rehli, M., and

Schneider-Brachert, W. (2004) Induction of maturation and cytokine release of human dendritic

cells by Helicobacter pylori. Infect Immun 72: 4416-4423.

Kranzer, K., Sollner, L., Aigner, M., Lehn, N., Deml, L., Rehli, M., and Schneider-Brachert, W.

(2005) Impact of Helicobacter pylori virulence factors and compounds on activation and

maturation of human dendritic cells. Infect Immun 73: 4180-4189.

Page 101: Ang Michelle 201006 MSc Thesis

88

Kusters, J. G., van Vliet, A. H., and Kuipers, E. J. (2006) Pathogenesis of Helicobacter pylori

infection. Clin Microbiol Rev 19: 449-490.

Lambeth, J. D. (2004) NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 4:

181-189.

Lee, H. K., Lund, J. M., Ramanathan, B., Mizushima, N., and Iwasaki, A. (3-9-2007)

Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science 315: 1398-1401.

Lenahan, C. and Avigan, D. (2006) Dendritic cell defects in patients with cancer: mechanisms

and significance. Breast Cancer Res 8: 101.

Lepper, P. M., Triantafilou, M., Schumann, C., Schneider, E. M., and Triantafilou, K. (2005)

Lipopolysaccharides from Helicobacter pylori can act as antagonists for Toll-like receptor 4. Cell

Microbiol 7: 519-528.

Levine, B. and Deretic, V. (2007) Unveiling the roles of autophagy in innate and adaptive

immunity. Nat Rev Immunol 7: 767-777.

Levine, B. and Klionsky, D. J. (2004) Development by self-digestion: molecular mechanisms

and biological functions of autophagy. Dev Cell 6: 463-477.

Levine, B. and Kroemer, G. (1-11-2008) Autophagy in the pathogenesis of disease. Cell 132: 27-

42.

Levy, D. E. and Darnell, J. E., Jr. (2002) Stats: transcriptional control and biological impact. Nat

Rev Mol Cell Biol 3: 651-662.

Lin, A., Schildknecht, A., Nguyen, L. T., and Ohashi, P. S. (1-4-2010a) Dendritic cells integrate

signals from the tumor microenvironment to modulate immunity and tumor growth. Immunol

Lett 127: 77-84.

Lin, A., Schildknecht, A., Nguyen, L. T., and Ohashi, P. S. (1-4-2010b) Dendritic cells integrate

signals from the tumor microenvironment to modulate immunity and tumor growth. Immunol

Lett 127: 77-84.

Lin, A., Schildknecht, A., Nguyen, L. T., and Ohashi, P. S. (1-4-2010c) Dendritic cells integrate

signals from the tumor microenvironment to modulate immunity and tumor growth. Immunol

Lett 127: 77-84.

Lin, T. and Bost, K. L. (9-3-2004) STAT3 activation in macrophages following infection with

Salmonella. Biochem Biophys Res Commun 321: 828-834.

Linden, S., Nordman, H., Hedenbro, J., Hurtig, M., Boren, T., and Carlstedt, I. (2002) Strain- and

blood group-dependent binding of Helicobacter pylori to human gastric MUC5AC glycoforms.

Gastroenterology 123: 1923-1930.

Page 102: Ang Michelle 201006 MSc Thesis

89

Liu, L., McBride, K. M., and Reich, N. C. (6-7-2005) STAT3 nuclear import is independent of

tyrosine phosphorylation and mediated by importin-alpha3. Proc Natl Acad Sci U S A 102: 8150-

8155.

Lochhead, P. and El Omar, E. M. (2007) Helicobacter pylori infection and gastric cancer. Best

Pract Res Clin Gastroenterol 21: 281-297.

Lu, Y. C., Yeh, W. C., and Ohashi, P. S. (2008) LPS/TLR4 signal transduction pathway.

Cytokine 42: 145-151.

Lunz, J. G., III, Specht, S. M., Murase, N., Isse, K., and Demetris, A. J. (2007) Gut-derived

commensal bacterial products inhibit liver dendritic cell maturation by stimulating hepatic

interleukin-6/signal transducer and activator of transcription 3 activity. Hepatology 46: 1946-

1959.

Marino, G., Salvador-Montoliu, N., Fueyo, A., Knecht, E., Mizushima, N., and Lopez-Otin, C.

(6-22-2007) Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient

in Atg4C/autophagin-3. J Biol Chem 282: 18573-18583.

Marnett, L. J. (2000) Oxyradicals and DNA damage. Carcinogenesis 21: 361-370.

Mashimo, M., Nishikawa, M., Higuchi, K., Hirose, M., Wei, Q., Haque, A., Sasaki, E., Shiba,

M., Tominaga, K., Watanabe, T., Fujiwara, Y., Arakawa, T., and Inoue, M. (2006) Production of

reactive oxygen species in peripheral blood is increased in individuals with Helicobacter pylori

infection and decreased after its eradication. Helicobacter 11: 266-271.

Mastroeni, P., Vazquez-Torres, A., Fang, F. C., Xu, Y., Khan, S., Hormaeche, C. E., and

Dougan, G. (7-17-2000) Antimicrobial actions of the NADPH phagocyte oxidase and inducible

nitric oxide synthase in experimental salmonellosis. II. Effects on microbial proliferation and

host survival in vivo. J Exp Med 192: 237-248.

Mathew, R., Karp, C. M., Beaudoin, B., Vuong, N., Chen, G., Chen, H. Y., Bray, K., Reddy, A.,

Bhanot, G., Gelinas, C., DiPaola, R. S., Karantza-Wadsworth, V., and White, E. (6-12-2009)

Autophagy suppresses tumorigenesis through elimination of p62. Cell 137: 1062-1075.

Meijer, A. J. and Codogno, P. (2006) Signalling and autophagy regulation in health, aging and

disease. Mol Aspects Med 27: 411-425.

Melillo, J. A., Song, L., Bhagat, G., Blazquez, A. B., Plumlee, C. R., Lee, C., Berin, C., Reizis,

B., and Schindler, C. (3-1-2010c) Dendritic Cell (DC)-Specific Targeting Reveals Stat3 as a

Negative Regulator of DC Function. J Immunol 184: 2638-2645.

Melillo, J. A., Song, L., Bhagat, G., Blazquez, A. B., Plumlee, C. R., Lee, C., Berin, C., Reizis,

B., and Schindler, C. (3-1-2010a) Dendritic Cell (DC)-Specific Targeting Reveals Stat3 as a

Negative Regulator of DC Function. J Immunol 184: 2638-2645.

Melillo, J. A., Song, L., Bhagat, G., Blazquez, A. B., Plumlee, C. R., Lee, C., Berin, C., Reizis,

B., and Schindler, C. (3-1-2010b) Dendritic Cell (DC)-Specific Targeting Reveals Stat3 as a

Negative Regulator of DC Function. J Immunol 184: 2638-2645.

Page 103: Ang Michelle 201006 MSc Thesis

90

Mimuro, H., Suzuki, T., Tanaka, J., Asahi, M., Haas, R., and Sasakawa, C. (2002) Grb2 is a key

mediator of helicobacter pylori CagA protein activities. Mol Cell 10: 745-755.

Molinari, M., Salio, M., Galli, C., Norais, N., Rappuoli, R., Lanzavecchia, A., and Montecucco,

C. (1-5-1998) Selective inhibition of Ii-dependent antigen presentation by Helicobacter pylori

toxin VacA. J Exp Med 187: 135-140.

Montecucco, C. and Rappuoli, R. (2001) Living dangerously: how Helicobacter pylori survives

in the human stomach. Nat Rev Mol Cell Biol 2: 457-466.

Morgan, C., Jenkins, G. J., Ashton, T., Griffiths, A. P., Baxter, J. N., Parry, E. M., and Parry, J.

M. (10-6-2003) Detection of p53 mutations in precancerous gastric tissue. Br J Cancer 89: 1314-

1319.

Murphy, M. P. (1-1-2009) How mitochondria produce reactive oxygen species. Biochem J 417:

1-13.

Narendra, D., Tanaka, A., Suen, D. F., and Youle, R. J. (12-1-2008) Parkin is recruited

selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183: 795-803.

Necchi, V., Manca, R., Ricci, V., and Solcia, E. (2009) Evidence for transepithelial dendritic

cells in human H. pylori active gastritis. Helicobacter 14: 208-222.

Obonyo, M., Cole, S. P., Datta, S. K., and Guiney, D. G. (2006) Evidence for interleukin-1-

independent stimulation of interleukin-12 and down-regulation by interleukin-10 in Helicobacter

pylori-infected murine dendritic cells deficient in the interleukin-1 receptor. FEMS Immunol Med

Microbiol 47: 414-419.

Odenbreit, S., Puls, J., Sedlmaier, B., Gerland, E., Fischer, W., and Haas, R. (2-25-2000)

Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion.

Science 287: 1497-1500.

Ohnishi, N., Yuasa, H., Tanaka, S., Sawa, H., Miura, M., Matsui, A., Higashi, H., Musashi, M.,

Iwabuchi, K., Suzuki, M., Yamada, G., Azuma, T., and Hatakeyama, M. (1-22-2008) Transgenic

expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in

mouse. Proc Natl Acad Sci U S A 105: 1003-1008.

Olzmann, J. A. and Chin, L. S. (1-1-2008) Parkin-mediated K63-linked polyubiquitination: a

signal for targeting misfolded proteins to the aggresome-autophagy pathway. Autophagy 4: 85-

87.

Orvedahl, A. and Levine, B. (2009) Eating the enemy within: autophagy in infectious diseases.

Cell Death Differ 16: 57-69.

Ostrand-Rosenberg, S. (2008) Immune surveillance: a balance between protumor and antitumor

immunity. Curr Opin Genet Dev 18: 11-18.

Page 104: Ang Michelle 201006 MSc Thesis

91

Palacino, J. J., Sagi, D., Goldberg, M. S., Krauss, S., Motz, C., Wacker, M., Klose, J., and Shen,

J. (4-30-2004) Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J Biol

Chem 279: 18614-18622.

Paludan, C., Schmid, D., Landthaler, M., Vockerodt, M., Kube, D., Tuschl, T., and Munz, C. (1-

28-2005) Endogenous MHC class II processing of a viral nuclear antigen after autophagy.

Science 307: 593-596.

Pan, J. S., Hong, M. Z., and Ren, J. L. (4-14-2009) Reactive oxygen species: a double-edged

sword in oncogenesis. World J Gastroenterol 15: 1702-1707.

Papini, E., de Bernard, M., Milia, E., Bugnoli, M., Zerial, M., Rappuoli, R., and Montecucco, C.

(10-11-1994) Cellular vacuoles induced by Helicobacter pylori originate from late endosomal

compartments. Proc Natl Acad Sci U S A 91: 9720-9724.

Parsonnet, J., Friedman, G. D., Orentreich, N., and Vogelman, H. (1997) Risk for gastric cancer

in people with CagA positive or CagA negative Helicobacter pylori infection. Gut 40: 297-301.

Peek, R. M., Jr. and Crabtree, J. E. (2006) Helicobacter infection and gastric neoplasia. J Pathol

208: 233-248.

Portal-Celhay, C. and Perez-Perez, G. I. (2006) Immune responses to Helicobacter pylori

colonization: mechanisms and clinical outcomes. Clin Sci (Lond) 110: 305-314.

Qu, X., Yu, J., Bhagat, G., Furuya, N., Hibshoosh, H., Troxel, A., Rosen, J., Eskelinen, E. L.,

Mizushima, N., Ohsumi, Y., Cattoretti, G., and Levine, B. (2003) Promotion of tumorigenesis by

heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest 112: 1809-1820.

Rad, R., Brenner, L., Krug, A., Voland, P., Mages, J., Lang, R., Schwendy, S., Reindl, W.,

Dossumbekova, A., Ballhorn, W., Wagner, H., Schmid, R. M., Bauer, S., and Prinz, C. (2007)

Toll-like receptor-dependent activation of antigen-presenting cells affects adaptive immunity to

Helicobacter pylori. Gastroenterology 133: 150-163.

Raju, D. and Jones, N. L. (2010) Methods to monitor autophagy in H. pylori vacuolating

cytotoxin A (VacA)-treated cells. Autophagy 6: 138-143.

Reggiori, F., Shintani, T., Nair, U., and Klionsky, D. J. (2005) Atg9 cycles between

mitochondria and the pre-autophagosomal structure in yeasts. Autophagy 1: 101-109.

Reich, N. C. and Liu, L. (2006) Tracking STAT nuclear traffic. Nat Rev Immunol 6: 602-612.

Romero-Adrian, T. B., Leal-Montiel, J., Monsalve-Castillo, F., Mengual-Moreno, E., McGregor,

E. G., Perini, L., and Antunez, A. (2010) Helicobacter pylori: bacterial factors and the role of

cytokines in the immune response. Curr Microbiol 60: 143-155.

Samavati, L., Rastogi, R., Du, W., Huttemann, M., Fite, A., and Franchi, L. (2009) STAT3

tyrosine phosphorylation is critical for interleukin 1 beta and interleukin-6 production in

response to lipopolysaccharide and live bacteria. Mol Immunol 46: 1867-1877.

Page 105: Ang Michelle 201006 MSc Thesis

92

Scherz-Shouval, R., Shvets, E., Fass, E., Shorer, H., Gil, L., and Elazar, Z. (4-4-2007) Reactive

oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO

J 26: 1749-1760.

Singh, M., Prasad, K. N., Saxena, A., and Yachha, S. K. (2006) Helicobacter pylori induces

apoptosis of T- and B-cell lines and translocates mitochondrial apoptosis-inducing factor to

nucleus. Curr Microbiol 52: 254-260.

Smith, M. F., Jr., Mitchell, A., Li, G., Ding, S., Fitzmaurice, A. M., Ryan, K., Crowe, S., and

Goldberg, J. B. (8-29-2003) Toll-like receptor (TLR) 2 and TLR5, but not TLR4, are required for

Helicobacter pylori-induced NF-kappa B activation and chemokine expression by epithelial

cells. J Biol Chem 278: 32552-32560.

Stax, A. M., Crul, C., Kamerling, S. W., Schlagwein, N., van der Geest, R. N., Woltman, A. M.,

and van Kooten, C. (2008) CD40L stimulation of rat dendritic cells specifically favors the IL-

12/IL-10 ratio resulting in a strong T cell stimulatory capacity. Mol Immunol 45: 2641-2650.

Steinman, R. M. and Banchereau, J. (9-27-2007) Taking dendritic cells into medicine. Nature

449: 419-426.

Sundrud, M. S., Torres, V. J., Unutmaz, D., and Cover, T. L. (5-18-2004) Inhibition of primary

human T cell proliferation by Helicobacter pylori vacuolating toxin (VacA) is independent of

VacA effects on IL-2 secretion. Proc Natl Acad Sci U S A 101: 7727-7732.

Szabo, I., Brutsche, S., Tombola, F., Moschioni, M., Satin, B., Telford, J. L., Rappuoli, R.,

Montecucco, C., Papini, E., and Zoratti, M. (10-15-1999) Formation of anion-selective channels

in the cell plasma membrane by the toxin VacA of Helicobacter pylori is required for its

biological activity. EMBO J 18: 5517-5527.

Takeshima, E., Tomimori, K., Takamatsu, R., Ishikawa, C., Kinjo, F., Hirayama, T., Fujita, J.,

and Mori, N. (2009) Helicobacter pylori VacA activates NF-kappaB in T cells via the classical

but not alternative pathway. Helicobacter 14: 271-279.

Tan, J. K. and O'Neill, H. C. (2005) Maturation requirements for dendritic cells in T cell

stimulation leading to tolerance versus immunity. J Leukoc Biol 78: 319-324.

Terebiznik, M. R., Raju, D., Vazquez, C. L., Torbricki, K., Kulkarni, R., Blanke, S. R.,

Yoshimori, T., Colombo, M. I., and Jones, N. L. (2009) Effect of Helicobacter pylori's

vacuolating cytotoxin on the autophagy pathway in gastric epithelial cells. Autophagy 5: 370-

379.

Terebiznik, M. R., Vazquez, C. L., Torbicki, K., Banks, D., Wang, T., Hong, W., Blanke, S. R.,

Colombo, M. I., and Jones, N. L. (2006) Helicobacter pylori VacA toxin promotes bacterial

intracellular survival in gastric epithelial cells. Infect Immun 74: 6599-6614.

Torres, V. J., VanCompernolle, S. E., Sundrud, M. S., Unutmaz, D., and Cover, T. L. (10-15-

2007) Helicobacter pylori vacuolating cytotoxin inhibits activation-induced proliferation of

human T and B lymphocyte subsets. J Immunol 179: 5433-5440.

Page 106: Ang Michelle 201006 MSc Thesis

93

Uno, K., Kato, K., Atsumi, T., Suzuki, T., Yoshitake, J., Morita, H., Ohara, S., Kotake, Y.,

Shimosegawa, T., and Yoshimura, T. (2007) Toll-like receptor (TLR) 2 induced through TLR4

signaling initiated by Helicobacter pylori cooperatively amplifies iNOS induction in gastric

epithelial cells. Am J Physiol Gastrointest Liver Physiol 293: G1004-G1012.

Vicari, A. P., Chiodoni, C., Vaure, C., Ait-Yahia, S., Dercamp, C., Matsos, F., Reynard, O.,

Taverne, C., Merle, P., Colombo, M. P., O'Garra, A., Trinchieri, G., and Caux, C. (8-19-2002)

Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide

and anti-interleukin 10 receptor antibody. J Exp Med 196: 541-549.

Wang, T., Niu, G., Kortylewski, M., Burdelya, L., Shain, K., Zhang, S., Bhattacharya, R.,

Gabrilovich, D., Heller, R., Coppola, D., Dalton, W., Jove, R., Pardoll, D., and Yu, H. (2004)

Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat

Med 10: 48-54.

Wang, Y. H., Wu, J. J., and Lei, H. Y. (2009) The autophagic induction in Helicobacter pylori-

infected macrophage. Exp Biol Med (Maywood ) 234: 171-180.

Weinberg, F. and Chandel, N. S. (2009) Reactive oxygen species-dependent signaling regulates

cancer. Cell Mol Life Sci 66: 3663-3673.

White, E., Karp, C., Strohecker, A. M., Guo, Y., and Mathew, R. (1-5-2010) Role of autophagy

in suppression of inflammation and cancer. Curr Opin Cell Biol.

Willhite, D. C. and Blanke, S. R. (2004a) Helicobacter pylori vacuolating cytotoxin enters cells,

localizes to the mitochondria, and induces mitochondrial membrane permeability changes

correlated to toxin channel activity. Cell Microbiol 6: 143-154.

Willhite, D. C. and Blanke, S. R. (2004b) Helicobacter pylori vacuolating cytotoxin enters cells,

localizes to the mitochondria, and induces mitochondrial membrane permeability changes

correlated to toxin channel activity. Cell Microbiol 6: 143-154.

Willhite, D. C., Cover, T. L., and Blanke, S. R. (11-28-2003) Cellular vacuolation and

mitochondrial cytochrome c release are independent outcomes of Helicobacter pylori vacuolating

cytotoxin activity that are each dependent on membrane channel formation. J Biol Chem 278:

48204-48209.

Wilson, K. T. and Crabtree, J. E. (2007) Immunology of Helicobacter pylori: insights into the

failure of the immune response and perspectives on vaccine studies. Gastroenterology 133: 288-

308.

Wu, S., Rhee, K. J., Albesiano, E., Rabizadeh, S., Wu, X., Yen, H. R., Huso, D. L., Brancati, F.

L., Wick, E., McAllister, F., Housseau, F., Pardoll, D. M., and Sears, C. L. (2009) A human

colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell

responses. Nat Med 15: 1016-1022.

Xie, Z. and Klionsky, D. J. (2007) Autophagosome formation: core machinery and adaptations.

Nat Cell Biol 9: 1102-1109.

Page 107: Ang Michelle 201006 MSc Thesis

94

Yakes, F. M. and Van Houten, B. (1-21-1997) Mitochondrial DNA damage is more extensive

and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc

Natl Acad Sci U S A 94: 514-519.

Yang, J. and Stark, G. R. (2008) Roles of unphosphorylated STATs in signaling. Cell Res 18:

443-451.

Yang, X. O., Panopoulos, A. D., Nurieva, R., Chang, S. H., Wang, D., Watowich, S. S., and

Dong, C. (3-30-2007) STAT3 regulates cytokine-mediated generation of inflammatory helper T

cells. J Biol Chem 282: 9358-9363.

Yorimitsu, T. and Klionsky, D. J. (2005) Autophagy: molecular machinery for self-eating. Cell

Death Differ 12 Suppl 2: 1542-1552.

Yue, Z., Jin, S., Yang, C., Levine, A. J., and Heintz, N. (12-9-2003) Beclin 1, an autophagy gene

essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl

Acad Sci U S A 100: 15077-15082.

Zhang, X., Yue, P., Fletcher, S., Zhao, W., Gunning, P. T., and Turkson, J. (1-11-2010) A novel

small-molecule disrupts Stat3 SH2 domain-phosphotyrosine interactions and Stat3-dependent

tumor processes. Biochem Pharmacol.

Zheng, P. Y. and Jones, N. L. (2003) Helicobacter pylori strains expressing the vacuolating

cytotoxin interrupt phagosome maturation in macrophages by recruiting and retaining TACO

(coronin 1) protein. Cell Microbiol 5: 25-40.

Zou, W. (2005) Immunosuppressive networks in the tumour environment and their therapeutic

relevance. Nat Rev Cancer 5: 263-274.