enhancing host immunity to avaian influenza virus …

115
Enhancing Host Immunity to Avian Influenza Virus using Toll-like Receptor Agonists in Chickens By Michael St. Paul A Thesis presented to The University of Guelph In partial fulfillment of requirements for the degree of Master of Science in Pathobiology Guelph, Ontario, Canada © Michael St. Paul, July, 2012

Upload: others

Post on 19-May-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

i

Enhancing Host Immunity to Avian Influenza Virus using Toll-like

Receptor Agonists in Chickens

By

Michael St. Paul

A Thesis

presented to

The University of Guelph

In partial fulfillment of requirements

for the degree of

Master of Science

in

Pathobiology

Guelph, Ontario, Canada

© Michael St. Paul, July, 2012

Page 2: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

ii

ABSTRACT

ENHANCING HOST IMMUNITY TO AVIAN INFLUENZA VIRUS USING

TOLL-LIKE RECEPTOR AGONISTS IN CHICKENS

Michael St. Paul Advisor:

University of Guelph, 2012 Professor S. Sharif

Toll-like receptors (TLRs) are evolutionarily conserved pattern recognition

receptors that mediate host-responses to pathogens. In mammals, TLR ligands promote

cellular activation and the production of cytokines. Several TLR ligands have been

employed prophylactically for the control of bacterial or viral diseases in the mouse

model. However, the TLR-mediated responses in chickens have not been well described.

Importantly, the utility of TLR agonists for the control of viral pathogens, such as avian

influenza virus (AIV), has not been fully explored in chickens. To this end, the studies

described in this thesis characterized the kinetics of in vivo responses in chickens to the

TLR4 ligand lipopolysaccharide (LPS) and the TLR21 ligand CpG ODN. It was

demonstrated that both of these ligands induced the up-regulation of several immune

system genes in the spleen, including those associated with pro-inflammatory and

antiviral responses, as well antigen presentation. By harnessing the immunostimulatory

properties of TLR ligands, it was also demonstrated that the prophylactic administration

of either poly I:C (a TLR3 ligand), LPS or CpG ODN may confer immunity to a low

pathogenic avian influenza virus, as determined by a reduction in both oropharyngeal and

cloacal virus shedding in infected birds. Furthermore, transcriptional analysis of genes in

the spleen and lungs identified interleukin (IL)-8, interferon (IFN)-α and IFN-γ as

Page 3: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

iii

correlates of immunity. In conclusion, TLR ligands may modulate several aspects of the

chicken immune system to induce an anti-viral state, thereby conferring immunity to

AIV.

Page 4: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

iv

ACKNOWLEDGEMENTS

First, I would like to thank my advisor Dr. Shayan Sharif. Your guidance,

mentoring and support have been invaluable in providing me with a foundation for a

successful career in immunology. I also greatly appreciate the fact that you were always

very supportive of my many side projects, even though a few of them were definitely out

in left field!

I would also like to thank my advisory committee members, Dr. Éva Nagy and

Dr. Mohamed Faizal Abdul-Careem, for their guidance and assistance.

I would like to thank all past and present members of the Sharif lab, especially

Sarah, Neda, Mallick, Leah, Kamran, Ian, Payvand, Lianne, Jen, Doug and Shahriar. I

greatly appreciate your assistance and help in vaccinating, infecting bleeding and

sampling the chickens. I am especially thankful to Sarah, Neda, Mallick and Ian, who

were always willing to help me vaccinate chickens, process samples, bleed chickens or

isolate splenocytes, even if it meant working well into a Friday night.

I would also like to thank my friends and colleagues in the Pathobiology

department and elsewhere. I would also like to thank Claudia for her endless

encouragement, patience and support. No matter what happened with my research, you

were always able to show me the bright side of things. I am truly grateful and

appreciative of all the help and support that you have provided me.

Lastly, I would like to thank my family. You have always been there to support

me during my studies and I could not have achieved this without you.

Page 5: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

v

TABLE OF CONTENTS

ACKNOWLEDGEMENTS……………………………………………………………IV

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

LIST OF TABLES…………………………………………………………………...VIII

LIST OF FIGURES…………………………………………………………………….IX

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

INTRODUCTION………………………………………………………………………1

CHAPTER 1: ……………………………………………………………………………3

Literature Review...........................................................................................................................3

Toll-like Receptors...............................................................................................................4

TLR Structure and Function................................................................................................6

Chicken TLR3......................................................................................................................7

Chicken TLR4......................................................................................................................9

Chicken TLR21..................................................................................................................10

Exploiting TLR Ligands to Enhance Host-Resistances to Pathogens ..............................12

TLR Ligands Demonstrate Adjuvant Potential..................................................................16

The Influenza Virus...........................................................................................................19

Viral Replication Cycle......................................................................................................19

Viral Pathogenesis.............................................................................................................21

Host-Responses to Influenza Virus in Mammals..............................................................22

Immune Responses to AIV in Chickens............................................................................25

Vaccination........................................................................................................................27

STATEMENT OF RATIONALE……………………………………………………...30

STATEMENT OF HYPOTHESIS…………………………………………………….31

EXPERIMENTAL APPROACH……………………………………………………...32

Objective 1.........................................................................................................................32

Key steps involved................................................................................................32

Objective 2.........................................................................................................................32

Key steps involved................................................................................................32

Page 6: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

vi

CHAPTER 2…………………………………………………………………………….34

In vivo administration of ligands for chicken Toll-like receptors 4 and 21 induce

the expression of immune system genes in the spleen………………………………...34

Abstract..................................................................................................................35

Introduction............................................................................................................36

Materials and Methods...........................................................................................39

Experimental Chickens and Housing...............................................................39

TLR Ligands....................................................................................................39

Experimental Design........................................................................................39

RNA Extraction and cDNA Synthesis.............................................................40

Real-time PCR.................................................................................................40

Data Analysis...................................................................................................41

Results....................................................................................................................42

Expression of cytokine transcripts in the spleen following LPS

treatment...........................................................................................................42

Expression of cytokine transcripts in the spleen following CpG and non-CpG

ODN treatment..................................................................................................44

Discussion..............................................................................................................49

CHAPTER 3…………………………………………………………………………….57

Prophylactic treatment with Toll-like receptor ligands enhances host immunity to

avian influenza virus in chickens………………………………………………………57

Abstract..................................................................................................................58

Introduction............................................................................................................59

Materials and Methods...........................................................................................61

Experimental Chickens and Housing...............................................................61

Avian Influenza Virus......................................................................................61

TLR Ligands....................................................................................................61

Experimental Design........................................................................................62

Virus Titration..................................................................................................62

RNA Extraction, cDNA Synthesis and real-time PCR....................................63

Data Analysis...................................................................................................63

Results....................................................................................................................65

Treatment with TLR ligands reduces oropharyngeal and cloacal virus

shedding ...........................................................................................................65

Page 7: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

vii

Correlates of immunity in the spleen and lungs of chickens treated with TLR

ligands ..............................................................................................................66

Discussion..............................................................................................................69

CHAPTER 4…………………………………………………………………………….79

General Discussion...........................................................................................................79

REFERENCES………………………………………………………………………….85

Page 8: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

viii

LIST OF TABLES

Chapter 1

Table 1 Chicken Toll-like receptors and their ligands…………...………………5

Table 2 Summary of TLR ligand-mediated protection against various

pathogens………………...………………………………………………16

Chapter 2

Table 3 Primer sequences and accession numbers used for quantitative RT-

PCR………………………………………………………………………41

Table 4 Summary of transcriptional responses in the spleen following LPS and

CpG ODN treatment……………………………………………………..48

Chapter 3

Table 5 Summary of virus shedding and transcript levels in the lungs on days 4

and 7 p.i………………………………………………………………….68

Page 9: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

ix

LIST OF FIGURES

Chapter 2

Figure 1 (A) IFN-α transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (B) IFN-γ transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (C) IL-13 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (D) IL-10 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (E) IL-1β transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (F) IL-8 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (G) MyD88 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (H) iNOS transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 1 (I) MHC II transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

100 μg or 500 μg of LPS ………………………………………………..55

Figure 2 (A) IFN-α transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN……………………….……………………56

Figure 2 (B) IFN-γ transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN……………………………………………56

Figure 2 (C) IL-13 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN…………………………………………….56

Figure 2 (D) IL-10 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN...………………………………………….56

Figure 2 (E) IL-1β transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN……………………………………………56

Page 10: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

x

Figure 2 (F) IL-8 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with 10

μg or 50 μg of CpG ODN...……………………………………………...56

Figure 2 (G) MyD88 transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN..…………………………………………..56

Figure 2 (H) iNOS transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN...………………………………………….56

Figure 2 (I) MHC II transcripts in the spleens of chickens at 2, 6, 12 and 24 h.p.i. with

10 μg or 50 μg of CpG ODN..………………………………………….56

Chapter 3

Figure 3 (A) Oropharyngeal virus shedding on day 4 post-infection............................74

Figure 3 (B) Cloacal virus shedding on day 4 post-infection……………………........74

Figure 3 (C) Cloacal virus shedding on day 7 post-infection……………....................74

Figure 4 (A) IFN-α transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i……………………………………………………………………75

Figure 4 (B) IFN-β transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i…………………………………………………………………….75

Figure 4 (C) IFN-γ transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i…………………………………………………………………….75

Figure 4 (D) OAS transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i…………………………………………………………………….75

Figure 5 (A) IL-1β transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i…………………………………………………………………….76

Figure 5 (B) IL-8 transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and 7

d.p.i………………………………………………………………………76

Figure 5 (C) IL-18 transcripts in the spleens of TLR ligand-treated chickens at 2, 4 and

7 d.p.i…………………………………………………………………….76

Figure 6 (A) IFN-α transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i………………………………………...…………………………….77

Figure 6 (B) IFN-β transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

Page 11: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

xi

d.p.i………………………………………………...……………………..77

Figure 6 (C) IFN-γ transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i……………………………………………………………………….77

Figure 6 (D) OAS transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i……………………………………………………………………….77

Figure 7 (A) IL-1β transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i……………………………………………………………………….78

Figure 7 (B) IL-8 transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i……………………………………………………………………….78

Figure 7 (C) IL-18 transcripts in the lungs of TLR ligand-treated chickens at 2, 4 and 7

d.p.i……………………………………………………………………….78

Page 12: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

xii

LIST OF ABBREVIATIONS

AIV Avian influenza virus

APC Antigen presenting cell

cDNA Complementary DNA

CD Cluster of differentiation

DC Dendritic cell

DMEM Dulbecco’s Modified Eagle Medium

DNA Deoxyribonucleic Acid

d.p.i. Days post-infection

HA Hemagglutinin

HPAI Highly pathogenic avian influenza virus

HPI Hours post-injection

IBV Infectious bronchitis virus

IFN Interferon

IL Interleukin

iNOS Inducible nitric oxide synthase

LPAI Low pathogenic avian influenza virus

LPS Lipopolysaccharide

M1 Matrix protein 1

M2 Matrix protein 2

MAP Mitogen-activated protein

MD-2 Myeloid differentiation protein-2

Mda5 Melanoma differentiation-associated gene 5

MDCK Madin Darby Canine Kidney Cell

MHC Major histocompatibility complex

mL Milliliter

MyD88 Myeloid Differentiation Factor 88

NA Neuraminidase

NDV Newcastle disease virus

NK Natural killer

NP Nucleoprotein

NS1 Non-structural protein 1

NS2 Non-structural protein 2

OAS Oligoadenylate synthetase

ODN Oligodeoxynucleotide

PA Polymerase acidic protein

PAMPs Pathogen associated molecular patterns

PB1 Polymerase basic protein 1

PB1-F2 Polymerase basic protein 1-F2

PB2 Polymerase basic protein 2

PBS Phosphate-buffered saline

PCR Polymerase chain reaction

PI Post-infection

Poly I:C Polyinosinic:polycytidylic acid

PRR Pattern recognition receptor

qRT-PCR Quantitative reverse-transcription PCR

Page 13: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

xiii

RIG-I Retinoic acid inducible gene I

RNA Ribonucleic acid

siRNA Short interfering RNA

TGF Transforming growth factor

TCID50 50% Tissue culture infectious dose

TH T-helper

TIR Toll-IL 1 Receptor

TLR Toll-like receptor

TNF Tumour necrosis factor

TRIF TIR-domain-containing adapter-inducing interferon- β

µL Microliter

WHO World Health Organization

Page 14: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

xiv

Declaration of work performed

The studies described in this thesis are original and the experiments were performed by

Michael St. Paul. Technical assistance was provided by in alphabetical order: Kamran

Haq, Amirul I. Mallick, Shahriar Orouji, Payvand Parvizi, Leah R. Read and Alexander

Ian Villanueva, with the intellectual assistance of Dr. Mohamed Faizal Abdul-Careem

and Dr. Éva Nagy. All work was carried out under the supervision of Dr. Shayan Sharif

at the Department of Pathobiology, University of Guelph.

Page 15: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

1

INTRODUCTION

Harnessing the power of the innate immune system holds promise in protecting

chickens against viral pathogens. Traditionally, the innate immune system is defined as

having several classic hallmarks, namely the potential to respond to a broad range of

pathogens immediately, as well as lacking specificity and immunological memory. As

such, the innate immune system serves as the first line of defence against pathogens and

microbes. This is largely achieved through a multilayered approach, consisting of

anatomical, physiological and cellular barriers (Kindt et al., 2006). For example,

anatomical barriers include the skin, and mucous membranes, while physiological

barriers include stomach acid, fever, complement, lysozyme and anti-microbial peptides.

However, if these two barriers are overcome, the innate immune system relies on the

cellular barrier, which is constituted by a variety of cell subsets including macrophages,

natural killer (NK) cells, heterophils/neutrophils, mast cells, basophils and eosinophils.

Together, these cell subsets protect the host through a variety of mechanisms, such as by

phagocytosis of foreign particles and microbes as well as through the production of

cytokines that may induce a pro-inflammatory state. Importantly, certain cells of the

innate immune system, such as macrophages, play a critical role in initiating and

modulating the adaptive immune response through antigen presentation. Adaptive

immune responses are mediated both by antibodies and cell-mediated mechanisms,

relying on B cells and T cells, respectively. The adaptive immune system is thought to

have specificity, diversity, self/non-self recognition and memory. However, recent

discoveries have clearly shown that members of the innate immune system may also

share at least some of the above features with the adaptive immune system.

Page 16: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

2

In mammals and chickens, there is a high degree of integration and cross-talk

between the innate and adaptive arms of the immune system. For example, cytokines

produced by dendritic cells, such as interleukin (IL)-12, and by mast cells or other cells,

such as IL-4, influence the polarization of T-helper (TH) cells into either a TH1 or TH2

phenotype, which are important for mediating responses against intracellular or

extracellular pathogens, respectively. Conversely, cytokines, such as interferon (IFN)-γ,

as well as signaling molecules, such as CD40 ligand, produced by T cells activate innate

immune system cells and enhance their effector functions (Buhtoiarov et al., 2005).

Together, this integration and crosstalk confers both flexibility and robustness to the

immune response, thereby conferring protection against a variety of pathogens.

Therefore, following the administration of compounds that activate the innate immune

system, the resulting cellular activation and cytokine production may be exploited to

protect chickens against viral pathogens, including avian influenza virus.

Page 17: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

3

CHAPTER 1:

Literature Review

Michael St. Paul

Department of Pathobiology, University of Guelph, Guelph, Ontario

Modified versions of the different sections of the Literature Review will be submitted for

publication:

St. Paul, M., Brisbin, J. and Sharif, S. Immunomodulatory properties of Toll-like receptor

ligands in chickens. To be submitted to Veterinary Immunology and Immunopathology

Page 18: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

4

Toll-Like Receptors

To identify potential invaders and discriminate self from non-self, cells of the

innate immune system rely on pattern recognition receptors (PRRs) to recognize and bind

conserved motifs found in bacteria and viruses, known as pathogen-associated molecular

patterns (PAMPs) (Medzhitov and Janeway, 1997). Although there are several different

types of PRRs, the one type that has been studied the most is the Toll-like receptor

(TLR). In 1996, it was found that the Toll receptor was required to protect Drosophila

from a fungal pathogen (Lemaitre et al., 1996). It was soon after this discovery that

Medzhitov and colleagues showed that this receptor is evolutionarily conserved, as

numerous TLRs were subsequently detected in humans, mice and chickens (Medzhitov et

al., 1997; Roach et al., 2005). To date, at least 12 TLRs have been identified in mice and

10 in chickens (Temperley et al., 2008). Together, these molecules mediate responses to

pathogens by binding different types of PAMPs. In humans and mice for example, TLRs

3 and 9 recognize double-stranded RNA and CpG DNA motifs, respectively, which are a

characteristic of certain viral and bacterial nucleic acids, while TLR4 recognizes

lipopolysaccharides (LPS) found in the cell wall of Gram-negative bacteria (Chow et al.,

1999; Akira and Takeda, 2004). Although mice and chickens share many TLR orthologs

including TLRs 3 and 4, chickens possess unique TLRs as well, namely TLRs 15 and 21.

While recognizing the same CpG DNA ligand as mammalian TLR9, TLR21 is

evolutionarily distinct from TLR9 and is thus classified as a separate TLR (Keestra et al.,

2010). Although most of the chicken TLRs are expressed on the cell surface, TLRs 3, 7,

and 21 are typically located intracellularly, and are recruited to the endosomes following

the endocytosis/phagocytosis of a pathogen (Keestra et al., 2010). This phenomenon may

allow for more frequent interactions with their ligands, which are all some type of nucleic

Page 19: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

5

acid, and as such are not exposed to TLRs until the pathogen has been digested in the

endosome. A summary of chicken TLRs and their ligands may be found in Table 1.

Table 1. Chicken Toll-like receptors and their ligands

TLR Structure

recognized

Agonist

TLR1a - -

TLR1b - -

TLR2a Bacterial cell

wall components Peptidoglycan

TLR2b - -

TLR3 Double stranded

RNA Poly I:C

TLR4 Bacterial cell

wall components LPS

TLR5 Bacterial

Flagellin Flagellin

TLR7 Single-stranded

RNA

Imiquimod,

Resiquimod

TLR15 Unknown

TLR21 CpG DNA CpG ODN

TLRs are expressed by a wide variety of cells including macrophages, dendritic

cells, neutrophils, NK cells and mast cells (Hopkins and Sriskandan, 2005). While largely

considered to be an innate receptor, recent evidence has also detected TLRs in lymphoid

cells, such as B cells, T cells and invariant natural killer T cells (Hanten et al., 2008).

Stimulation of TLRs on these cell subsets results in cellular activation, maintenance of

memory cells and exertion of effector functions (Kulkarni et al., 2010). In chickens,

TLRs have been detected in several cell subsets including in heterophils (Kogut et al.,

2005b), monocytes and macrophages (He et al., 2006), as well as in B cells and T cells

(Iqbal et al., 2005). Furthermore, chicken TLR transcripts have been detected in several

Page 20: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

6

tissues including in the spleen, the bursa of Fabricius, the lungs, and several regions of

the gastrointestinal tract such as the duodenum and jejunum (Iqbal et al., 2005).

TLR Structure and Function

TLRs are transmembrane proteins consisting of three domains: the extracellular

domain containing leucine rich repeats, the transmembrane domain, and the intracellular

Toll/IL-1 receptor (TIR) domain (Akira et al., 2001). Following binding of a ligand to the

extracellular domain, the TIR domain recruits adaptor molecules, two key ones being

Myeloid Differentiation Factor 88 (MyD88) and TIR-domain-containing adapter-

inducing interferon- β (TRIF) (Moynagh, 2005). In mammals, it has been shown that all

TLRs, except TLRs 3 and 4, utilize MyD88 dependent pathways exclusively, recruiting

solely MyD88 and not TRIF. Moreover, TLR3 utilizes the MyD88-independent pathway,

recruiting TRIF and not MyD88, while TLR4 utilizes both the MyD88-dependent and

independent pathways. Nonetheless both pathways are similar in that they lead to the

downstream activation of mitogen-activated protein (MAP) kinases and nuclear factor

(NF)-κB (Kawai and Akira, 2010). NF-κB is a transcription factor, which when activated,

translocates into the nucleus promoting transcription of pro-inflammatory genes,

including IL-1β, IL-6 and tumor necrosis factor (TNF)-α, in addition to the up-regulation

of co-stimulatory molecules and the maturation of antigen presentation cells (Akira and

Hoshino, 2003).

In chickens, the MyD88 and TRIF genes have both been identified and cloned by

our group (Wheaton et al., 2007). Similar to mammals, it appears that many of the

chicken TLRs signal through the MyD88-dependent pathway (Keestra and van Putten,

Page 21: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

7

2008; Ciraci and Lamont, 2011). Although it has not yet been demonstrated whether

chicken TLR3 signals through the TRIF pathway, there is some debate as to whether

TLR4 signals through both pathways. A study by Keestra and colleagues (2008)

suggested that MyD88-independent signaling does not occur with chicken TLR4, as

demonstrated by the lack of type I interferon up-regulation in response to LPS. In

contrast, it has recently been proposed that TLR4 may in fact signal through the TRIF

pathway, as chicken macrophages produce type I IFNs in response to LPS (Lian et al.,

2012). Taken together, early evidence suggests that both mammalian and chicken TLRs

utilize many of the same signaling pathways, however more studies are needed to

determine the extent of the utilization of the TRIF pathway in chickens.

As all of the studies that are described in the present thesis involved the use of

ligands for TLRs 3, 4 and 21, the immunomodulatory properties of only these TLRs in

chickens will be discussed in detail.

Chicken TLR3

Transcripts for chicken TLR3 have been detected in several tissues, such as the

spleen and lungs, as well as in several cell subsets including heterophils, B cells and T

cells (Iqbal et al., 2005). As mentioned earlier, the PAMP associated with TLR3 is

double-stranded RNA, and the most well studied TLR3 ligand used in mammals is the

synthetic double-stranded RNA, polyinodinic:polycytidylic acid (Poly I:C). Indeed, it

appears that poly I:C is also stimulatory for chicken TLR3 as well (Schwarz et al., 2007),

and its immunomodulatory effects have been documented in previous studies. In

heterophils for example, treatment with poly I:C significantly increased degranulation,

Page 22: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

8

while down-regulating the production of the pro-inflammatory cytokines IL-1β, IL-6 and

IL-8 (Kogut et al., 2005b). Similarly, chicken macrophages and fibroblasts treated with

poly I:C up-regulated TLR3 transcripts as well as transcripts for IFN-α (Karpala et al.,

2008). The study by Karpala and colleagues (2008) also found that using RNA

interference targeting TLR3 in chicken fibroblasts reduced the amount of IFN-α produced

in response to poly I:C. As such, it is evident that poly I:C is recognized by chicken

TLR3 and mediates cellular activation and the production of cytokines. It should be noted

that poly I:C is not the only immunostimulatory synthetic double-stranded RNA in

chickens. Studies in our lab have compared the efficacy of the dsRNA ligands β-

Galactosidase 728, poly U:G, SARS coronavirus-targeting dsRNA molecule, poly I:C

and the dsRNA molecule targeting enhanced green fluorescent protein, in up-regulating

TLR3 transcripts as well as certain cytokines, such as the type I IFNs, in chicken

splenocytes (Villanueva et al., 2011). All of these molecules were indeed determined to

be immunostimulatory, with poly I:C inducing the most robust and rapid cytokine

response. In contrast, the responses to poly U:G were lower compared to responses to

poly I:C, however the duration of this response was more prolonged when compared to

that of poly I:C-induced responses.

In addition to in vitro studies, our group has also demonstrated that poly I:C is

immunostimulatory for chickens in vivo (Parvizi et al., 2012). In a recent study, poly I:C

was administered through three different routes: aerosol, intra-airsac and intramuscular

(i.m.), and subsequently gene expression in both the spleen and lung was examined. It

was found that poly I:C administered through the i.m. route induced the greatest up-

regulation of IFN-α and IFN-β in both the spleen and lungs (Parvizi et al. 2012). Taken

Page 23: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

9

together, it is clear that poly I:C is indeed immunostimulatory for chickens, and responses

to poly I:C are mediated in part by chicken TLR3.

Chicken TLR4

The chicken TLR4 gene was sequenced in 2003 by Leveque and colleagues,

where they also noted that allelic variation in TLR4 was linked to susceptibility to

Salmonella enterica serovar Typhimurium. It was later confirmed that similar to

mammals, LPS is the ligand for chicken TLR4 and binding of LPS in chickens is

mediated by the formation of the TLR4/myeloid differentiation protein-2 (MD-2)

complex, as well as by the accessory proteins lipopolysaccharide binding protein and

CD14 (Kogut et al., 2005a; Keestra and van Putten, 2008). Transcripts for TLR4 have

been detected in numerous cell subsets, such as in macrophages, heterophils,

thrombocytes and B cells, as well as in tissues including the spleen and bursa of Fabricius

(Iqbal et al., 2005), where they help to mediate host-responses to several pathogens

including species of Salmonella (Leveque et al., 2003; Abasht et al., 2008; Chaussé et al.,

2011) and Clostridium perfringens (Lu et al., 2009).

The in vitro responses to LPS have been characterized in several immune system

cell subsets in chickens. For example, our group has shown that chicken B cells respond

to LPS by up-regulating several immune system genes including TLR4 transcripts and

CD164 (Sarson et al., 2007). Furthermore, thrombocytes up-regulate several transcripts

associated with pro-inflammatory responses including the cytokines IL-1β and IL-6, as

well as increasing the production of cyclooxygenase-2 and prostaglandin E2 in response

to LPS (Ferdous et al., 2008; Scott and Owens, 2008). In addition, LPS treatment has

Page 24: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

10

been shown to induce the production of nitrite in avian macrophages (He, Genovese, et

al., 2011) and promote the respiratory burst and degranulation of heterophils (Kogut et

al., 2005).

Although well studied in vitro, only a few studies have characterized the

immunomodulatory effects of LPS in vivo. Previously, it has been shown that LPS

administered intravenously increases the concentration of pro-inflammatory cytokines in

the serum at 1 hour post-treatment, in addition to increasing the numbers of circulating

leukocytes, such as heterophils (Boever et al., 2009). In addition, intravenous

administration of LPS has been shown to induce the up-regulation of some immune

system genes in the spleen, namely IFN-γ, IL-6, IL-8 and IL-15, at 2 hours post-treatment

(Sijben et al., 2003).

Chicken TLR21

Chicken TLR21 is a unique TLR that functions similar to mammalian TLR9, as

they both mediate responses to CpG oligodeoxynucleotides (ODNs) (Keestra et al.,

2010). Similar to mammals, it has been shown in chickens that certain sequences of the

ODNs are more immunostimulatory than others. Depending on the sequence of the ODN,

it may be grouped into one of 3 classes, A, B or C (Vollmer et al., 2004). Class A ODNs

are highly immunostimulatory to plasmacytoid dendritic cells (pDC) and thereby induce

the production of IFN-α (Krug et al., 2001). In contrast, class B ODNs weakly induce

IFN-α production, but are highly immunostimulatory for B cells, thereby resulting in

enhanced IFN-γ production (Krug et al., 2001). Lastly, class C ODNs are a mixture of

Class A and B motifs, thereby moderately stimulating both pDCs and B cells. In

Page 25: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

11

chickens, the vast majority of previous studies have used the class B CpG ODN 2007, as

it has been shown to be one of the most immunostimulatory ODN sequences for chicken

TLR21 (He et al., 2003; Brownlie et al., 2009).

Transcripts for TLR21 have been detected in several immune system cell subsets

and tissues including B cells, macrophages and heterophils, as well as in the spleen and

bursa of Fabricius (Brownlie et al., 2009; Wattrang, 2009; Keestra et al., 2010).

Responses mediated by TLR21 include cellular activation and proliferation of chicken B

cells (Wattrang, 2009) and the production of IL-6 in splenocytes (Jenkins et al., 2009).

Furthermore, CpG ODNs up-regulate pro-inflammatory cytokines in chicken

macrophages and enhance their nitric oxide production (He et al., 2003; Brownlie et al.,

2009). In an in vivo study by Patel and colleagues (2008), it was revealed that after

treatment with CpG, some immune system genes, including IFN-γ and IL-10, were up-

regulated in both the spleen and bursa of Fabricius.

Responses to TLR21 ligands may be synergistically enhanced when administered

with ligands for TLR3. This has been demonstrated in chicken monocytes that were

stimulated with CpG ODN and poly I:C, as they demonstrated a significant increase in

effector functions as well as a 100 to 1000-fold greater up-regulation of IFN-γ transcripts

when compared to treatment with either ligand alone (He et al., 2007a, 2012).

Page 26: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

12

Exploiting TLR ligands to enhance host-resistances to pathogens

Owing in part to their immunostimulatory properties, TLR ligands have been

employed prophylactically as stand-alone agents to protect against several pathogens in

mammals. In humans for example, ligands for TLRs 3, 7 and 9 are the subject of

numerous clinical trials aimed at protecting against viral infections such as treating

hepatitis C virus, genital herpes and human papilloma viruses (Kanzler et al., 2007). In

addition to humans, a similar phenomenon has also been observed in mice. It was found

that the prophylactic administration of TLR ligands may also confer immunity to several

types of pathogens, such as influenza virus, as several studies have shown that poly I:C,

LPS and CpG ODN are all effective in protecting mice against lethal influenza virus

challenges (Wong et al., 1995, 2009; Shinya et al., 2012). From this, it is clear that the

prophylactic administration of TLR ligands has the potential to protect mammals against

a variety of pathogens,

Based on their success in mammals, several studies over the past decade have

been aimed at investigating whether this phenomenon may be extended to chickens.

Indeed, it appears that this is the case, as it has been shown that TLR ligands can protect

chickens from bacterial, viral or parasitic organisms as outlined in Table 2. For the most

part, the majority of TLR ligands used in this context are those which bind to TLRs 3, 7

or 21. As all three of these TLRs recognize viral PAMPs, these TLRs modulate the

immune system towards an anti-viral state mediated in part through the production of a

robust type I or type II interferon response.

Type I interferons play an important role in mediating protection against many

viral pathogens, such as avian influenza virus (AIV). In addition to up-regulating the

Page 27: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

13

interferon inducible genes (ex.2’-5’ oligoadenylate synthetase), these cytokines inhibit

viral transcription and translation, promote the apoptosis of infected cells (Samuel, 2001).

In this regard, ligands that induce a robust IFN-α and IFN-β response make them an

attractive strategy for enhancing immunity against AIV. Although signaling through

either the MyD88 or TRIF pathways culminate in type I IFN production, a greater

magnitude of type I IFN responses are typically associated with the TRIF pathway. To

this end, TLR3 ligands have been used in vitro and in vivo to protect chickens and

chicken cell populations against both low and high pathogenic AIV. For example, it was

demonstrated by Stewart et al. (2011) that incorporating a 5’-UGUGU-3’ motif into small

interfering RNAs (siRNA) inhibited the replication of highly pathogenic AIV H5N1 in

chicken macrophages. In this scenario, it is likely that the protection conferred by TLR3

ligands was mediated through two mechanisms: 1) the direct activation of macrophages,

thereby enhancing their effector functions such as phagocytic capabilities and the

production of nitric oxide, and 2) the production of type I IFN. In fact, type I interferons,

especially IFN-α, are particularly efficacious in enhancing host immunity to AIV, as IFN-

α acts to contain the spread of the virus through pre-activating uninfected cells, thereby

reducing their susceptibility to infection (Jiang et al., 2011; Meng et al., 2011).

As mentioned previously, the induction of a robust type I IFN response is not

limited to only those ligands that signal through the TRIF pathway. In particular, TLR7

ligands have been shown to induce the up-regulation of these cytokines in chicken

splenocytes (Stewart et al., 2011a). To this end, it was demonstrated that stimulating

chicken macrophages in vitro with the TLR7 ligand loxoribine significantly inhibited

PR8 influenza virus replication and reduced virus titre in the supernatant (Stewart et al.,

Page 28: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

14

2011a). Moreover, it was found that injecting loxoribine into emrbyonated chicken eggs

24 hours prior to infection with PR8 significantly reduced the amount of virus found in

the allantoic fluid. As such, it is clear that IFN-α, and the TLR ligands that induce its up-

regulation, present themselves as an attractive prophylactic strategy against viral

infections. However, as the studies done so far in chickens involving AIV were

conducted in vitro, it remains to be determined whether similar protective effects occur in

vivo.

It is not only type I interferons that demonstrate anti-viral properties, cytokines

associated with TH1 responses, such as IFN-γ, may also induce an anti-viral state. This is

mediated in part through the activation of NK cells and CD8+ T cells. Together, these

cell subsets exert their anti-viral effects partly through the targeted release of cytotoxic

granules towards infected cells, thereby promoting their apoptosis. Moreover, IFN-γ may

also up-regulate the interferon inducible genes, thereby protecting uninfected cells. In this

regard, CpG ODN, which induces a TH1-like response in chickens (Patel et al., 2008), has

been shown to be efficacious in protecting against several viral pathogens, such as

infectious bronchitis virus (IBV) (Dar et al., 2009b). Indeed, as shown by Dar and

colleagues (2009), a reduction in IBV load was correlated with an up-regulation of IFN-γ

and the interferon inducible gene 2’-5’ oligoadenylate synthetase (OAS), which initiates

the RNase L pathway and promotes the cleavage of viral RNA.

It is also important to note that the efficacy of CpG ODN prophylaxis is not just

limited to viruses, as it may also confer immunity against bacterial and parasitic

pathogens. A study by Dalloul et al. (2004) demonstrated that CpG ODN administered

intravenously decreases the amount of oocyst shedding in chickens infected with the

Page 29: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

15

parasite Eimeria, while at the same time improving weight gain in chickens. Moreover,

several studies have focused on the protective effects of CpG ODN against bacterial

pathogens such as Escherichia coli in chickens (Gomis et al., 2003, 2004). This is

despite the fact that E. coli is not an intracellular pathogen and therefore the activation of

CD8+ T cells and the induction of many of the interferon inducible genes are not as

relevant in this context. This suggests that other TLR21 mediated mechanisms may be

involved, such as the direct activation of monocytes and heterophils, thereby increasing

their phagocytic uptake of bacteria. Another possibility is through the direct activation of

B cells, as CpG ODN is highly immunostimulatory for chicken B cells and enhances their

effector functions including cytokine production and the up-regulation of both major

histocompatibility complex (MHC) class II and the relevant co-stimulatory molecules

(Wattrang, 2009; St. Paul et al., unpublished results). As such, it is clear that CpG ODN

has the potential to confer immunity against a broad spectrum of pathogens.

Page 30: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

16

Table 2. Summary of TLR ligand-mediated protection against various pathogens.

TLR Agonist Pathogen Outcome Reference

TLR3

5’-UGUGU-3’ AIV (H5N1)

Reduced virus

replication in

vitro

(Stewart et al., 2011b)

Poly I:C Avian reovirus

Reduced

plaque

formation in

infected CEFs

(Marcus and Sekellick,

2001)

Poly I:C Newcastle disease

virus

Reduced

plaque

formation in

infected CEFs

(Marcus and Sekellick,

2001)

TLR5 Flagellin Salmonella

enteritidis

Reduced

mortality (Genovese et al., 2007)

TLR7 Loxoribine PR8 influenza virus

(H1N1)

Reduced virus

replication in

vitro and in ovo

(Stewart et al., 2011a)

TLR21

CpG ODN Eimeria acervulina

Decreased

oocyst

shedding

(Dalloul et al., 2004)

CpG ODN Infectious bronchitis

virus

Reduced virus

replication in

chick embryos

(Dar et al., 2009b)

CpG ODN Escherichia coli Reduced

mortality

(Gomis et al., 2004;

Taghavi et al., 2009)

CpG ODN Salmonella spp.

Reduced

mortality or

bacterial load

(He et al., 2005;

Taghavi et al., 2008;

Mackinnon et al., 2009)

TLR Ligands demonstrate adjuvant potential

The protective benefits of TLR ligands are not limited to prophylactic

administration prior to a microbial challenge, as many ligands have shown promise as

vaccine adjuvants to enhance antibody production and T cell responses. In this regard,

CpG ODN has been the most widely used TLR ligand in chickens to enhance the

immunogenicity of vaccines against several pathogens including AIV. Our group has

shown that CpG ODN demonstrates superior adjuvant potential compared to recombinant

Page 31: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

17

IFN-γ or diluent, in eliciting antibody-mediated and cell-mediated responses to avian

influenza virosomes (Mallick et al., 2011). A similar finding has been extended towards

vaccines against the highly pathogenic H5N1, as Wang and colleagues (2009) found that

adding CpG ODN to the oil and water emulsion significantly enhanced antibody

responses and hemagglutination inhibition (HI) antibody titres, while increasing the

amount of serum IFN-γ. Although the exact immunological basis behind CpG ODN’s

superior adjuvant potential has not been well described in chickens, it is hypothesized

that this may be a result of 1) a robust IFN-γ response, which acts to mature antigen

presenting cells and enhance their antigen presenting capabilities (Patel et al., 2008) and

2) the direct activation of B cells, thereby promoting the up-regulating of co-stimulatory

molecules, MHC II and cytokine transcripts (St. Paul et al., submitted).

In addition to CpG ODN, a few of the other TLR ligands have been shown to

possess adjuvant potential in chickens, such as poly I:C, Pam3CSK4, LPS and flagellin

(Erhard et al., 2000, Chaung et al., 2011, Parvizi et al., submitted). In mice, it has been

well established that the TLR5 ligand flagellin enhances mucosal immune responses and

this property has been exploited to enhance the immunogenicity of vaccines against

pathogens which enter through mucosal surfaces, such as influenza virus (Wang et al.,

2010). A similar phenomenon has recently been demonstrated in chickens, as Chaung et

al. (2011) discovered that an adjuvant consisting of flagellin mixed with aluminum gel

significantly enhanced secretory IgA antibodies following intranasal vaccination with an

inactivated H5N2 AIV. The increase in secretory IgA may be attributed in part to the

TH2-like response observed in chicken splenocytes following treatment with flagellin (St.

Paul et al., submitted).

Page 32: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

18

Not all TLR ligands can serve as an adjuvant to enhance antibody responses, as it

has been shown that LPS may in fact decrease antibody responses. In a study by

Parmentier et al. (2004) administration of lipoteichoic acids (LTA) 24 hours prior to

immunization with keyhole limpet hemocyanin (KLH) significantly augmented anti-KLH

antibodies. In contrast, pre-treatment with LPS significantly down-regulated the antibody

responses to KLH when compared to PBS-treated control birds. A similar phenomenon

was observed following an intratracheal administration of Newcastle disease virus (NDV)

co-encapsulated in a liposome with LPS, as liposomes without LPS induced significantly

higher antibody titres and increased survival rates after a single immunization (Tseng et

al., 2009). In contrast, Parmentier et al. (2008) found that antibody responses were

significantly increased following an intratracheal immunization of chickens with HSA

along with LPS as an adjuvant. The mechanisms behind this discrepancy is not known at

this time, and it may be related to the source of LPS, the dose and the route of

administration, as all three of these factors may in fact influence the expression of

immune system genes in chicken lymphoid tissues (Pulendran et al., 2001; Parvizi et al.,

2012). Taken together, these studies demonstrate the potential for LPS to both augment

or decrease antibody responses, and future studies may be aimed at optimizing LPS

formulations as adjuvant

Having a proven track record of enhancing immune response to avian influenza

vaccines, as well as enhancing immunity to avian influenza virus in vitro, administration

of TLR ligands prophylactically may serve as a novel approach to protect chickens

against AIV in vivo.

Page 33: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

19

Influenza Virus

Influenza virus is an enveloped, negative-sense single-stranded RNA virus

belonging to the family Orthomyxoviridae. There are three types of influenza viruses,

each classified to a corresponding genus of the same name: Type A, Type B and Type C

(Suarez and Schultz-Cherry, 2000). Unlike types B and C, which are confined mostly to

humans, influenza A viruses have the largest host range, infecting a variety of animal

species including swine, humans, poultry and waterfowl (Suarez and Schultz-Cherry,

2000). Influenza A viruses are further classified into subtypes according to the presence

of the surface glycoproteins hemagglutinin (HA) and neuraminidase (NA). To date, 16

HA and 9 NA subtypes have been identified (Rappuoli and Del Giudice, 2008).

The influenza genome is composed of 8 segments that encode 11 viral genes

(Samji, 2009). The glycoproteins HA and NA, are found on the surface of the virion,

while the matrix protein 2 (M2) is a proton channel integrated into the viral envelope,

which is composed of the host’s plasma membrane. Approximately 80% of the surface

proteins are HA, with 17% being NA and the remainder being M2 (Samji, 2009). The

internal proteins of the virus consist of the polymerase acidic protein (PA), polymerase

basic protein 1 (PB1), polymerase basic protein 2 (PB2) and polymerase basic protein 1-

F2 (PB1-F2), the nucleoprotein (NP), the matrix protein 1 (M1) and the non-structural

protein 2 (NS2). The viral genome also encodes the non-structural protein 1 (NS1), which

is produced in large quantities but not packaged into the virion.

Viral Replication Cycle

Page 34: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

20

The replication cycle of influenza virus begins with the HA of the virion binding

to sialic acid expressed on the surface of host cells. In humans, cells in the respiratory

tract contain α(2,6) linkages between the sialic acid and the carbohydrate galactose,

while in avian species, cells contain α(2,3) linkages (Rappuoli and Del Giudice, 2008).

The determining factor for host and cell tropism of an influenza virus strain depends on

the sialic acid specificity of the HA expressed. For example, avian influenza viruses have

a preference for α(2,3) linkages, while human viruses have an affinity for α(2,6) linkages.

Interestingly, porcine cells express both α(2,3) and α(2,6) linkages, making them ideal

‘mixing pots’ for virus recombination (Rappuoli and Del Giudice, 2008). Nevertheless,

following the binding of HA to sialic acid, the virus is endocytosed via receptor-mediated

endocytosis by the host cell, enclosing the virus in an endosome. The low pH of the

endosome (pH 5-6) promotes a conformational change in the HA exposing HA1 and

HA2, the latter of which mediates fusion between the host membrane and viral envelope.

In addition, the low pH also allows for passage of ions into the virion via M2, thereby

acidifying the viral core and promoting the release of the viral ribonucleoproteins

(vRNPs) into the cytoplasm of the host cell (Samji, 2009).

The vRNPs is composed of the viral RNA in conjunction with NP and the

polymerase proteins PB1, PB2 and PA (Samji, 2009). Together, these proteins provide

the vRNP with nuclear localization signals and facilitate its entry into the nucleus. In the

nucleus, the viral RNA is used as a template to generate positive sense RNA, which is

mediated by the viral polymerases. Some positive sense viral RNA strands are

polyadenylated and serve as a template for the translation of viral proteins, while other

full-length positive sense viral RNA strands are not polyadenylated and act as a template

Page 35: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

21

for the replication of new viral RNA strands to be packaged into newly forming virions.

The viral protein M1 then mediates virion assembly, and the progeny virions exit the cell

by budding from the host cell plasma membrane (Samji, 2009). To prevent re-

agglutination of the progeny virus to the host cell, the NA on the progeny virus

subsequently cleaves off sialic acid from the host cell.

Viral Pathogenesis

Based on their pathogenesis, avian influenza viruses can be classified into one of

two types, highly pathogenic avian influenza (HPAI) or low pathogenic avian influenza

(LPAI). While the majority of subtypes are classified as LPAI, a few strains belonging to

either the H5 or H7 subtypes are considered to be highly pathogenic, such as H5N1. The

major difference between LPAI and HPAI is the ability of the virus to replicate locally or

systemically, respectively (Lee and Suarez, 2005). This is achieved largely by mutations

in HPAI viruses allowing the HA to be cleaved by a wider range of proteases found

throughout the host tissues. Furthermore, these proteases may be expressed in the cell

cytoplasm therefore cleaving HA of virus progeny before they are released from the cell,

resulting in an increase in their cell tropism (Suarez, 2010). As such, following a HPAI

challenge, infectious virus can be detected in a variety of tissues including in the brain,

heart and blood vessels (Brown et al., 1992), while LPAI tend to remain confined to the

mucosal surfaces including the lungs and the gastrointestinal tract (Slemons and Swayne,

2010). The clinical signs associated with LPAI infection in chickens are mild and often

remain undetected, and include ruffled feathers as well as a drop in egg production

(Reynolds, 2006). However, infection with HPAI strains results in a rapid spread within

Page 36: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

22

and between flocks, leading to severe respiratory and neurological signs, multi-organ

failure and 90-100% mortality (Chmielewski and Swayne, 2011). Therefore, in order for

an avian influenza virus subtype to be officially classified as HPAI according to the

United States Department of Agriculture, the virus must be lethal to 75% of the infected

birds upon intravenous administration of the virus.

Host Responses to Influenza Virus in Mammals

TLRs and other PRRs play an important role in mediating host-responses to

influenza virus. Following infection of murine lung epithelial cells, influenza virus is

recognized by the cytosolic PRR retinoic acid inducible gene (RIG)-1, which binds RNA

containing 5’ triphosphate ends (Le Goffic et al., 2007). Furthermore, it has been

hypothesized that epithelial TLR3 recognizes the dsRNA intermediary occurring during

viral replication, however the extent of contributions made through TLR3 stimulation to

host-responses against influenza virus is unclear and is subject to debate (Guillot et al.,

2005; Le Goffic et al., 2007). Nevertheless, TLR7 is thought to be the main TLR

responsible for initiating host-responses to influenza virus through binding its ssRNA

(Ehrhardt et al., 2010). As TLR7 is located intracellularly in macrophages and dendritic

cells, this TLR encounters its ligand upon endocytosis of whole influenza virions or of

virally infected dead epithelial cells. Here, TLR7 stimulation facilitates the maturation of

dendritic cells, which subsequently migrate to the draining lymph node to present virus

antigens to T cells (Dzopalic et al., 2010). In addition, macrophages, along with infected

epithelial cells, produce certain cytokines and chemokines, including the pro-

inflammatory cytokines TNF-α, IL-1β, IL-6, as well as the anti-viral type-1 IFNs (IFN-

Page 37: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

23

α/β), which serve to recruit and activate other leukocytes in addition to controlling the

early spread of the virus through activating epithelial cells as well as promoting the

apoptosis of infected epithelial cells (Peiris et al., 2010).

Within 48 h following infection, natural killer cells and neutrophils are recruited

into the lungs to control early viral replication (Rappuoli and Del Giudice, 2008). Here,

NK cells play an important role in controlling the virus during the early stages of

infection through the production of IFN-γ and the lysis of virally infected cells. As such,

it is not surprising that mice depleted of NK cells have an increased virus load following

an influenza challenge ,when compared to mice that have normal NK cell numbers and

functions (Nogusa et al., 2008). In addition to NK cells, neutrophils also contribute to the

containment of the virus during the early stages of infection. This is achieved in part

through the phagocytosis of virions as well as producing anti-microbial peptides that

contribute to the early host defenses against influenza virus (Gaudreault and Gosselin,

2008). Neutrophils also produce cytokines in response to influenza virus (Wang et al.,

2008) and help sustain subsequent CD8+ T cell responses (Tate et al., 2011).

Furthermore, mice depleted of neutrophils have a higher titre of influenza virus in the

lung and demonstrate signs of an increased severity of disease (Tate et al., 2008).

T cells activated in the draining lymph node by dendritic cells play an important

role in viral clearance. Activated CD4+ T cells are able to provide the necessary help for

antigen-specific B cells through TCR stimulation and the production of cytokines and

CD40L, providing the signals required to differentiate B cells into plasma cells (Thomas

et al., 2006). Antibodies secreted from these plasma cells are generally targeted toward

the HA and NA surface glycoproteins and are important for immunity to infection, as

Page 38: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

24

well as neutralizing extracellular virions to help contain the spread (Rappuoli and Del

Giudice, 2008). In addition to antibody-mediated responses, CD8+ cytotoxic T

lymphocytes also contribute to containment and clearance of the virus, where they arrive

in the lungs on days 5-7 post-infection and kill virus- infected cells. To this end, it has

been shown that adoptive transfer of CD8+ T cells from mice vaccinated with influenza

virus into naive mice may confer immunity to these naïve mice when challenged with the

same strain of virus (Ulmer et al., 1998).

During the course of the immune response a wide array of cytokines are

produced, with one group of essential cytokines being the type I interferons. Therefore, to

antagonize the host IFN response, influenza viruses produce NS1 protein to prevent its

viral RNA from binding to intracellular sensors, among other functions (Kochs et al.,

2007). Moreover, NS1 aids in the pathogenesis of influenza viruses through the inhibition

of the interferon inducible genes, which hinder viral replication and promote the early

containment of the virus (Min and Krug, 2006). The importance of NS1 in the

pathogenesis of highly pathogenic AIV is evident through its identification as a key

virulence factor of H5N1 HPAI, thereby further supporting the significance of type I

IFNs in host immunity to influenza viruses (Li et al., 2006). However, even though type I

IFNs and other cytokines in the milieu work to protect the host, deleterious responses can

occur following infection with highly pathogenic strains of influenza, in what is

described as a ‘cytokine storm’. The cytokine storm leads to prolonged and extensive

inflammation and recruitment of immune cells, in addition to edema, leading to

respiratory distress and potential death of the host (Rappuoli and Del Giudice, 2008).

Page 39: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

25

Immune responses to AIV in chickens

There is a wealth of knowledge about mechanisms of immunity against influenza

viruses in mammals. However, little is known about these mechanisms in chickens.

Moreover, the importance of the contributions made by certain chicken cell subsets,

PRRs and cytokines has not been thoroughly investigated, partly due to the lack of

reagents. However, some aspects of the host-response towards AIV in chickens have

been elucidated, and for the most part are similar to what occurs in mammals.

The initial site of virus replication in chickens for both LPAI and HPAI viruses is

in the nasal cavity and lungs. Here, both types of viruses promote necrosis of the

parabronchial epithelium, edema and fibrin exudation in airways and air capillaries, with

the pathological lesions induced by HPAI infections being more severe (Degen et al.,

2006; Pantin-Jackwood and Swayne, 2009). During this time, both types of AIV are

detected by host pattern recognition receptors, namely TLRs and melanoma

differentiation-associated gene 5 (Mda5), but not RIG-I, as this PRR has not been

identified in chickens (Karpala et al., 2011b). Nevertheless, stimulation of PRRs initiates

the host immune response, mediated in part through the production of chemokines and

cytokines (Karpala et al., 2011b; Liniger et al., 2012). These cytokines and chemokines

serve to recruit and promote the influx of the early innate responders into the lungs, such

as macrophages and monocytes (Rebel et al., 2011). It is thought that these cell subsets

phagocytose virus-infected cells and process viral antigens for presentation to T cells, as

well as produce cytokines to contain the spread of the virus. These cytokines include type

I interferons, which act to up-regulate the interferon inducible genes, among other

functions. The importance of these interferon inducible genes was observed in chickens

Page 40: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

26

that had a variant of the myxomavirus-resistance (Mx) gene. The protein encoded by the

Mx gene in these chickens did not demonstrate anti-viral activities, and as a result, these

chickens had a higher virus load and mortality upon AIV infection, when compared to

those with the functional protein (Ewald et al., 2011). In addition to Mx, it was

determined through microarray analysis that another major interferon inducible gene, 2’-

5’ OAS, correlates with immunity to AIV in the lungs (Uchida et al., 2012). As such, it is

not surprising that IFN-α pre-treatment may protect chickens from AIV, again mediated

in part through the up-regulation of both Mx and 2’-5’ OAS (Jiang et al., 2011; Meng et

al., 2011).

In addition to type I IFNs, other cytokines and immune system genes are up-

regulated in the lungs of AIV-infected chickens. These include the pro-inflammatory

cytokines such as IL-6 as well as inducible nitric oxide synthase, which may both be

attributed to the influx of activated macrophages (Degen et al., 2006; Burggraaf et al.,

2011). Importantly, cytokines are produced in the lung that may bias the immune

response towards a TH1-phenotype, as demonstrated by the up-regulation of both IFN-γ

and IL-12, and not IL-4 (Karpala et al., 2011a). As a result, this leads to a robust CD8+ T

cell response and an influx of CD8α+ into the lungs of infected chickens (Rebel et al.,

2011). In conjunction with CTLs, anti-influenza antibody responses are detected at 1

week post-challenge, with antibody-secreting cells found in the lungs, spleen and bone

marrow (De Geus et al., 2011). Importantly, it appears that the CTL response is specific

against highly conserved epitopes spread across several influenza strains, raising the

possibility that the CTLs may be cross-protective (Seo and Webster, 2001; Haghighi et

al., 2009). Indeed, this is the case, as it was found that the adoptive transfer of H9N2

Page 41: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

27

specific CTLs into naïve chickens protected them from a lethal challenge with H5N1

(Seo and Webster, 2001). Similarly, it was found that chickens depleted of H9N2 specific

CD8+ T cells, but not CD4+ T cells, died as a result of lethal challenge with HPAI H5N1

(Seo et al., 2002). As such, it is clear that both antibody-mediated and cell-mediated

responses promote immunity against the virus.

Following the initial replication in the respiratory tract, both LPAI and HPAI

viruses spread to other organs. In the case of LPAI, this spread is limited to the

gastrointestinal tract. Here the host responds through up-regulating transcripts for TLRs

and cytokines such as IFN-β and IFN-γ, but not IFN-α (Nang et al., 2011). In contrast,

HPAI disseminate from the respiratory tract to the intestines as well as to several other

organs including the brain, kidneys, pancreas and the heart. As a result, the host-

responses to HPAIV occur in these tissues as well. For example, in the brain, a significant

up-regulation of heat shock proteins and chemokines such as IL-8, were observed, likely

serving to induce a pro-inflammatory response and promote the recruitment of leukocytes

(Balasubramaniam et al., 2012). However, the pro-inflammatory response occurring in

the brain and other organs results in immune-mediated pathological reactions, thereby

exacerbating the signs of disease. As a result, necrosis and apoptosis of cells in the brain,

as well as in the other organs lead to multi-organ failure, haemorrhage, edema and

thrombosis and finally death (Pantin-Jackwood and Swayne, 2009).

Vaccination

Poultry in regions where H5N1 is endemic are being vaccinated as a part of the

strategy to contain and limit the spread of the virus. The desired role of an ideal vaccine

Page 42: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

28

is to prevent or reduce infection and disease, to reduce the amount of virus being shed, to

allow easy differentiation between infected and vaccinated chickens, and to raise the

minimum threshold of exposure required for infection (Lee and Suarez, 2005). Currently,

two types of vaccines are being used commercially: inactivated whole virion oil-in water

emulsion vaccines, and a fowlpox recombinant vaccine (Capua and Marangon, 2006).

Conventional inactivated vaccines typically induce a predominant antibody-mediated

response with antibodies specific towards surface HA and NA, while recombinant

fowlpox vaccine induces both antibody-mediated and cell-mediated immune response

(Lee and Suarez, 2005; Haghihghi et al., 2010). However, despite eliciting antibodies and

T cells specific against HA and NA, current vaccines do not prevent infection and thus do

not completely eliminate shedding of the virus into the environment (Swayne, 2009).

To address the limited efficacy in reducing virus shedding, new types of vaccine

platforms are currently being developed, with one approach being influenza virosomes.

Influenza virosomes are reconstituted virus particles that are devoid of nucleic acids and

some of the viral protein, although they contain surface HA and NA. As a result, these

virosomes may bind to, and fuse with, the host cell membrane. Indeed, vaccination with

virosomes derived from the LPAI H4N6 has been shown to induce robust antibody-

mediated and cell-mediated immune responses against H4N6 virus, thereby reducing the

viral load in experimentally infected chickens (Mallick et al., 2011, 2012). However, it

remains to be elucidated if the virosome vaccine platform may protect chickens against

HPAIV.

In addition to virosomes, another promising AI vaccine platform is DNA

vaccines. Previously, it has been shown that DNA vaccines may induce neutralizing

Page 43: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

29

antibodies against highly pathogenic H5N1 and LPAI H5N2 in chickens, and

subsequently protect them from mortality and signs of disease (Kodihalli et al., 1997;

Jiang et al., 2007b; Rao et al., 2008). In addition, it was found that the antibody response

elicited by DNA vaccines may be broadly neutralizing against heterologous subtypes as

determined by a virus neutralization assay (Rao et al., 2008). However, one potential

drawback to DNA vaccines is the requirement of a gene gun in order to induce a more

robust immune response when compared to i.m. injection as determined by Rao and

colleagues (2008), and thus may serve as potential barrier for their widespread adoption.

In summary, it is clear that AIV vaccines may limit the spread of the virus and

protect chickens from mortality. However, existing commercial vaccines do not eliminate

virus shedding into the environment from infected birds. Although potentially promising

new vaccine platforms are under development, it remains to be determined how

efficacious they are outside of an experimental setting. As such, it is clear that novel

approaches should be identified to reduce the virus load and shedding in chickens

infected with AIV, with one potential approach being the prophylactic use of TLR

agonists.

Page 44: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

30

STATEMENT OF RATIONALE

It is evident that Toll-like receptor ligands modulate several aspects of the chicken

immune system by enhancing cell effector functions and inducing the production of

cytokines. These cytokines include the pro-inflammatory IL-1β and IL-8, and more

importantly, those associated with antiviral responses, namely type I IFNs. However,

although the TLR-mediated responses have been well studied in vitro, few studies have

examined how TLR ligands may modulate the chicken immune system in vivo.

Secondly, the prophylactic administration of TLR ligands in chickens has been

shown to enhance immunity to several types of pathogens, including E. coli. Moreover,

TLR ligands have been shown to inhibit avian influenza virus replication and reduce

virus load in vitro and in ovo. However, it remains to be elucidated as to whether a

similar phenomenon occurs in vivo,

Page 45: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

31

STATEMENT OF HYPOTHESIS

In the studies described in the present thesis, it was hypothesized that in vivo

administration of TLR ligands influences the expression of immune system genes in the

spleen. These genes likely include those associated with pro-inflammatory responses,

antigen presentation as well as anti-viral activity. Secondly, it was hypothesized that

when administered prophylactically, prior to challenge with a low pathogenicity avian

influenza virus, TLR ligands confer immunity to the host and significantly reduce virus

shedding. It was also hypothesized that TLR ligands elicit immunity against AIV that is

mediated by cytokines associated with anti-viral and TH1 responses, such as the type I

IFNs and IFN-γ.

Page 46: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

32

EXPERIMENTAL APPROACH

Objective 1:

To elucidate the mechanisms through which TLR4 and TLR21 stimulation modulates the

chicken immune system in vivo.

Key steps involved

Administered both a low and high dose of the TLR4 ligand LPS and the TLR21

ligand CpG ODN intramuscularly. Control groups received non-CpG ODN or

PBS.

Spleen tissue was collected at 2, 6, 12 and 24 hours post-treatment from 6

chickens in each group for RNA extraction and cDNA synthesis.

The relative expression of several immune system genes was quantified using

real-time PCR

Objective 2:

To identify a TLR agonist that enhances host-immunity to avian influenza virus when

administered prophylactically.

Key steps involved

Administered both a low and high dose of the TLR3 ligand poly I:C, the TLR4

ligand LPS, and the TLR21 ligand CpG ODN intramuscularly. One control group

received non-CpG ODN while the other two received PBS.

Twenty-four hours post-injection, chickens were infected with the low-pathogenic

AIV H4N6. One of the PBS-treated control groups remained un-infected.

Oropharyngeal and cloacal swabs were collected on days 4 and 7 post-infection.

Page 47: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

33

Spleen and lung tissue were collected on days 2, 4 and 7 post-infection for RNA

extraction and cDNA synthesis.

Virus shedding was assessed by titrating swab fluid in MDCK cells.

Potential correlates of immunity in the spleen and lung of TLR-ligand treated

birds were identified and quantified using real-time PCR.

Page 48: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

34

CHAPTER 2:

In vivo administration of ligands for chicken Toll-like receptors 4 and 21 induces the

expression of immune system genes in the spleen

Michael St. Paul a, Amirul I. Mallick

ab, Kamran Haq

a, Shahriar Orouji

a, Mohamed

Faizal Abdul-Careem c and

Shayan Sharif

a*.

a Department of Pathobiology, Ontario, Veterinary College, University of Guelph,

Guelph, Ontario, Canada, N1G 2W1.

b Present address: Department of Imaging Research, Sunnybrook Research Institute,

Sunnybrook Health Science Centre, University of Toronto, 2075 Bayview Avenue,

Toronto, Ontario, Canada, M4N 3M5.

c Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada, T2N

4N1.

*To whom correspondence should be addressed:

Dr. Shayan Sharif, Department of Pathobiology, Ontario Veterinary College, University

of Guelph, Guelph, Ontario, N1G 2W1, Canada.

Tel: 1 (519) 824-4120 ext. 54641, Fax: 1(519) 824-5930 Email: [email protected]

Published in Veterinary Immunology and Immunopathology 2011; 144 (3-4): 228-37

Page 49: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

35

Abstract

Toll-like receptors (TLRs) are a group of conserved proteins that play an

important role in pathogen recognition in addition to the initiation and regulation of

innate and adaptive immune responses. To date, several TLRs have been identified in

chickens, each recognizing different ligands. TLR stimulation in chickens has been

shown to play a role in host-responses to pathogens. However, the mechanisms through

which TLRs modulate the chicken immune system have not been well examined. The

present study was conducted to characterize the kinetics of responses to TLR4 and

TLR21 stimulation in chickens following intramuscular injections of their corresponding

ligands, lipopolysaccharide (LPS) and CpG oligodeoxynucleotides (ODNs), respectively.

To this end, relative expression of cytokine genes in the spleen was determined at 2, 6, 12

and 24 hours after injection of TLR ligands. The results indicated that LPS strongly

induced the up-regulation of some immune system genes early on in the response to

treatment, including interferon (IFN)-γ, interleukin (IL)-10, and IL-1β. Furthermore,

treatment with CpG ODN promoted the up-regulation of major histocompatibility

complex (MHC)-II, IFN-γ and IL-10. The response to CpG ODN appeared to be

somewhat delayed compared to the response to LPS. Moreover, we found a significant

increase in IFN-α gene expression in response to LPS but not CpG ODNs. Future studies

may be aimed to further characterize the molecular mechanisms of TLR activation in

chickens or to exploit TLR agonists as vaccine adjuvants.

Page 50: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

36

Introduction

Toll-like receptors (TLR) are highly conserved ligand binding proteins present in

numerous species including humans, mice and chickens (Roach et al., 2005). Being a

pattern recognition receptor (PRR), TLRs are responsible for the recognition of

conserved motifs found on bacteria, fungi, parasites and viruses known as pathogen

associated molecular patterns (PAMPS) (Medzhitov, 2001). In mammals, TLR4

recognizes lipopolysaccharide (LPS) found in the outer membrane of Gram-negative

bacteria (Chow et al., 1999), whereas TLR9 recognizes CpG DNA motifs found in

certain bacterial and viral nucleic acids (Akira and Takeda, 2004). Several TLRs have

been identified in chickens to date, with many of them being homologous to mammalian

TLRs (Fukui et al., 2001; Leveque et al., 2003; Iqbal et al., 2005; Temperley et al., 2008;

Keestra et al., 2010).

Host responses elicited by interactions between TLRs and their ligands are

achieved through the differential recruitment of adaptor molecules by the intracellular

domain of TLRs, an example of which is myeloid differentiation factor 88 (MyD88).

MyD88 recruits additional adaptor molecules, which subsequently lead to the

downstream activation of mitogen-activated protein kinases and transcription factor

nuclear factor (NF)-kappaB (Wesche et al., 1997). NF-kappaB induces the transcription

of pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6 and tumour necrosis

factor (TNF)-α, and promotes the transcription of co-stimulatory molecules and the

maturation of antigen presentation cells (APCs) (Akira and Hoshino, 2003). The MyD88-

dependent pathway is used by all of the mammalian TLRs except for TLR3, which

signals through the MyD88-independent pathway instead.

Page 51: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

37

The chicken TLR4 is an orthologue of mammalian TLR4, thereby binding LPS

following the formation of the TLR4/myeloid differentiation protein-2 (MD-2) complex

(Leveque et al., 2003; Keestra and Van Putten, 2008). Previously, the expression of

chicken TLR4 has been detected in many cell subsets, such as macrophages and B cells,

as well as in tissues including the spleen and bursa of Fabricius (Iqbal et al., 2005). LPS

treatment has been shown to induce the production of nitrite in avian macrophages (He et

al., 2011a) and promote the respiratory burst and degranulation of heterophils (Kogut et

al., 2005b). Moreover, LPS has previously been shown to induce the up-regulation of

cytokines, such as interferon (IFN)-γ in chicken B cells (Sarson et al., 2007) and pro-

inflammatory cytokines in the spleen (Sijben et al., 2003) and serum (Boever et al.,

2009). Furthermore, the possible role of TLR4 in eliciting host immune responses to

pathogens such as species of Salmonella (Leveque et al., 2003; Abasht et al., 2008;

Chaussé et al., 2011) and Clostridium perfringens (Lu et al., 2009) has been reported.

In mammals, responses to CpG ODN are largely mediated through TLR9.

However unlike TLR4, no orthologue of TLR9 has been identified in chickens to date.

Despite that, early research by Vleugels and colleagues (2002) established that CpG

ODN are immunostimulatory for chickens, while Gomis and colleagues (2003) identified

their immunoprotective nature. Further studies identified TLR21 as the TLR that interacts

with CpG ODN (Brownlie et al., 2009; Keestra et al., 2010). It has been demonstrated

that TLR21 may share a similar ligand specificity to that of TLR9, as the responses to a

given ODN may be influenced by the sequences flanking the CpG motif (He et al., 2003;

Ciraci and Lamont, 2011). In this regard, the class B CpG ODN, which preferentially

activates mammalian B cells in an IFN-α independent manner, has been shown to

Page 52: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

38

promote the proliferation of chicken B cells (Wattrang, 2009), and up-regulation of

cytokines such as IL-10 in the bursa of Fabricius (Patel et al., 2008) and IL-6 in

splenocytes (Jenkins et al., 2009). Furthermore, prophylactic treatment of chickens with

class B CpG ODN has been shown to enhance host immunity to numerous pathogens

such as Escherichia coli (Gomis et al., 2003; Gomis et al., 2004) and avian influenza

virus (St. Paul et al., in preparation), in addition to enhancing the immunogenicity of

avian-vaccines when administered as an adjuvant (Ameiss et al., 2006; Mallick et al.,

2011).

Although it is thought that stimulation with TLR4 and 21 agonists will promote a

pro-inflammatory response and up-regulation of IFNs as previously observed in vitro,

few studies have examined the kinetics of host responses in chickens to TLR stimulation

in vivo (Sijben et al., 2003; Patel et al., 2008). Therefore, the present study was an

attempt to understand and elucidate the mechanisms through which TLR4 and TLR21

stimulation modulates the chicken immune system. This was done by transcriptional

analysis of several immune system genes that represent various aspects of host responses

to TLR ligands. The results indicated that when TLRs 4 and 21 were stimulated, they

induced a dose-dependent up-regulation of IFN-γ, IL-8, inducible nitric oxide synthase

(iNOS), IL-10, MyD88, IL-1β and MHC-II, and in the case of TLR21, a down-regulation

of IL-13.

Page 53: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

39

Materials and Methods

Experimental chickens and housing

One-day-old broiler chickens (n=144) were procured from Maple Leaf Hatchery

(Hamburg, Ontario, Canada), and housed in the isolation facility of the Ontario

Veterinary College, University of Guelph (Guelph, ON). This research was approved by

the University of Guelph Animal Care Committee and complied with the guidelines of

the Canadian Council on Animal Care.

TLR ligands

Lipopolysaccharide (LPS) from Escherichia coli 0111:B4 was purchased from Sigma-

Aldrich-Canada (Oakville, ON), while synthetic class B CpG ODN 2007 [5’-

TCGTCGTTGTCGTTTTGTCGTT-3’] and non-CpG ODN [5’-

TGCTGCTTGTGCTTTTGTGCTT-3’] (Brownlie et al., 2009) were purchased from

Eurofins MWG Operon (Ebersberg, GER). All of the ligands used were re-suspended in

sterile phosphate buffered saline (PBS, pH 7.4).

Experimental design

Fourteen-day-old chickens were randomly divided into 6 groups of 24 birds each.

Chickens were immunized intramuscularly (i.m) in the pectoral muscle with either a high

or low dose of LPS or CpG ODNs, while two control groups received either non-CpG

ODNs or PBS. The “LPS low” and “LPS high” groups received 100 μg or 500 μg of LPS,

respectively, while the groups “CpG low” and “CpG high” received a 10 μg or 50 μg of

CpG ODN 2007, respectively. The two control groups received 50 μg of non-CpG ODNs

Page 54: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

40

or PBS. Doses were selected based on previous experiments in chickens involving these

TLR agonists (Gomis et al., 2004; Parvizi et al., manuscript in preparation). Sampling

was done at 2, 6, 12 and 24 hours post injection, and at each time point, 6 chickens from

each group were euthanized and spleen tissue was stored in RNAlater (Qiagen, Valencia,

CA) for RNA extraction. The spleen is a secondary lymphoid organ and was selected as

the sampling tissue as previous studies have demonstrated responses in the spleen

following i.m. injection of TLR agonists (Patel et al., 2008).

RNA extraction and cDNA synthesis

Total RNA was extracted from 50 mg of spleen tissue using TRIzol® (Invitrogen,

Carlsbad, CA) according to the manufacturer’s protocol. Following extraction, 10 μg of

RNA was treated with DNA Free® (Ambion, Austin, TX) DNAse, and 1 μg of purified

RNA was reverse transcribed to cDNA using Superscript® II First Strand Synthesis kit

(Invitrogen, Carlsbad, CA) and oligo-dT primers, according to the manufacturers

recommended protocol. The resulting cDNA was subsequently diluted 1:10 in DEPC

treated water.

Real-time PCR

Quantitative real-time PCR using SYBR Green was performed on diluted cDNA using

the LightCycler® 480 II (Roche Diagnostics GmbH, Mannheim, GER) as previously

described (Villanueva et al., 2011). Briefly, each reaction involved a pre-incubation at

95°C for 10 min, followed by 45 cycles of 95 °C for 10 min, 55 °C -64 °C (TA as per

primer) for 5 s, and elongation at 72 °C for 10 s. Subsequent melt curve analysis was

performed by heating to 95°C for 10 s, cooling to 65°C for 1 minute, and heating to

Page 55: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

41

97°C. Primers were synthesized by Sigma-Aldrich-Canada (Oakville, ON), and their

specific sequences, references and accession numbers are outlined in Table 3.

Data Analysis

Relative expression levels of all genes were calculated relative to the housekeeping gene

β-actin based on the formula developed by Pfaffl (Pfaffl, 2001). Gene expression was

displayed as fold changes relative to the PBS-treated control group. Fold changes,

standard error and statistical significance were calculated using the software REST 2009

(Qiagen, Valencia, CA) (Pfaffl et al., 2002). The software REST 2009 employs random

relocation and bootstrapping techniques to test for statistical significance between

treatment groups and the PBS-treated control group. Results were considered statistically

significant if p ≤ 0.05 (*) and p ≤ 0.01 (**).

Table 3. Primer sequences and accession numbers used for quantitative RT-PCR.

Target

Gene

Primer Sequence Accession

Number

Reference

MHC II F: 5’-CCACGGACGTGATGCAGAAC-3’

R: 5’-ACCGCGCAGGAACACGAAGA-3’ 113206149 (Thanthrige-Don et al.,

2010)

IFN-α F: 5’-ATCCTGCTGCTCACGCTCCTTCT-3’

R: 5’-GGTGTTGCTGGTGTCCAGGATG-3’ AB021154 (Villanueva et al., 2011)

IFN-γ F: 5’-ACACTGACAAGTCAAAGCCGCACA-3’

R: 5’-AGTCGTTCATCGGGAGCTTGGC-3’ X99774 (Brisbin et al., 2010)

IL-1β F: 5’-GTGAGGCTCAACATTGCGCTGTA-3’

R: 5’-TGTCCAGGCGGTAGAAGATGAAG-3’ Y15006 (Abdul-Careem et al.,

2009)

IL-8 F: 5’-CCAAGCACACCTCTCTTCCA-3’

R: 5’-GCAAGGTAGGACGCTGGTAA-3’ AJ009800 (Abdul-Careem et al.,

2009)

IL-10 F: 5’-AGCAGATCAAGGAGACGTTC-3’

R: 5’-ATCAGCAGGTACTCCTCGAT-3’ AJ621614 (Haq et al., 2010)

IL-13 F: 5’-ACTTGTCCAAGCTGAAGCTGTC-3’

R: 5’-TCTTGCAGTCGGTCATGTTGTC-3’ AJ621250 (Abdul-Careem et al.,

2008b)

iNOS F: 5’-GGCAGCAGCGTCTCTATGACTTG-3’

R: 5’-GACTTTAGGCTGCCCAGGTTG-3’ NM 204961 (Abdul-Careem et al.,

2007)

MyD88 F: 5’-AGCGTGGAGGAGGACTGCAAGAAG-3’

R: 5’-CCGATCAAACACACACAGCTTCAG-3’ NM

001030962

(Villanueva et al., 2011)

β-Actin F: 5’-CAACACAGTGCTGTCTGGTGGTA-3’

R: 5’-ATCGTACTCCTGCTTGCTGATCC-3’ X00182 (Haq et al., 2010)

Page 56: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

42

Results

Expression of cytokine transcripts in the spleen following LPS treatment

The changes in gene expression levels among the chickens treated with different

doses of LPS are expressed as fold changes relative to the PBS treated control group

(Figure 1).

To shed some light on the early type I interferon response, we quantified

transcripts of IFN-α. IFN-α transcripts increased by 2.06 fold in chickens receiving the

low dose (100 μg) LPS treatment, whereas these transcripts rose by 4.04 fold (p ≤ 0.05) at

2 hours post injection (h.p.i) in those receiving the high dose (500 μg) treatment (Figure

1A). By 6 h.p.i., transcript levels in both treatment groups were not statistically different

from the control group.

To assess cytokines associated with the modulation and regulation of the immune

response, we examined IFN-γ, IL-13 and IL-10, which are associated with T-helper

(TH)1, TH2 and anti-inflammatory responses, respectively. Treatment with high dose or

low dose of LPS resulted in a 236.65 (p ≤ 0.01) and 188.55 (p ≤ 0.01) fold increase of

IFN-γ transcripts, respectively, in the spleen at 2 h.p.i. (Figure 1B). From 6 h.p.i., the

transcript levels of IFN-γ for both the high and low dose groups decreased, with the high

dose having a 1.64 fold increase (p ≤ 0.05) at this time point. There was no statistically

significant difference in either group when compared to the control group at 12 and 24

h.p.i. Transcript levels of IL-13 were increased by 2.67 fold (p ≤ 0.05) in only the high

dose treatment group at 2 h.p.i. (Figure 1C). However, at 6 h.p.i, both the high and low

dose LPS treated groups had a 2.21 fold (p ≤ 0.01) and 2.30 fold (p ≤ 0.05) decrease in

Page 57: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

43

IL-13 transcripts, which remained reduced at both 12 and 24 h.p.i.. IL-10 transcripts were

significantly up-regulated at 2 h.p.i., with a 61.95 (p ≤ 0.01) and 32.58 (p ≤ 0.05) fold

increase, for the high and low dose treated groups, respectively (Figure 1D). By 6 h.p.i.,

transcript levels remained elevated with a 7.31 fold (p ≤ 0.01) increase for the high LPS

dose treatment, and a 3.12 fold (p ≤ 0.01) increase for the low LPS dose treatment. For

the low dose group, the transcript levels were not statistically different from those of the

control group at 12 h.p.i., in contrast to the high dose treated group whose IL-10

transcripts remained higher compared to the control group (1.91 fold, p ≤ 0.01).

However, by 24 h.p.i., transcript levels in the high dose treated group were subsequently

down-regulated by 2.42 fold (p ≤ 0.01).

To gain some insight into the pro-inflammatory response, we quantified

transcripts of two pro-inflammatory cytokines, IL-1β and IL-8. IL-1β transcripts were

significantly increased by 74.68 (p ≤ 0.01) and 54.72 (p ≤ 0.01) fold at 2 h.p.i. following

high and low dose LPS treatments, respectively (Figure 1E). Levels of IL-1β transcripts

declined by 6 h.p.i to 3.35 fold (p ≤ 0.01) for the high dose LPS treatment group, and

3.21 fold (p ≤ 0.01) for the low dose LPS treatment group. Gene expression remained

significantly higher in the high dose group at 24 h.p.i., 2.15 fold higher when compared

to controls (p ≤ 0.05). IL-8 transcripts were increased by 25.84 (p ≤ 0.01) and 21.92 (p ≤

0.01) fold, at 2 h.p.i. for both low and high dose LPS treatments, respectively (Figure 1F).

At 24 h.p.i., IL-8 transcripts present in the spleen of the low dose treated chickens were

not statistically different from those in the control group, though the high dose treated

group still had a 2.87 (p ≤ 0.01) fold increase in IL-8 transcripts.

Page 58: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

44

Lastly, we examined the transcriptional responses of relevant proteins and

transcription factors which are suggestive of TLR stimulation and APC activation.

MyD88 transcripts were significantly up-regulated by 4.30 fold (p ≤ 0.05) at 2 h.p.i.

following the high dose LPS treatment, while the low dose LPS treated chickens showed

a 3.75 fold increase (p ≤ 0.05) (Figure 1G). At 6 h.p.i., transcript levels of MyD88 in both

treatments groups were not statistically different from the PBS treated control group.

Furthermore, iNOS transcripts were significantly up-regulated at 2 h.p.i. by 18.01 (p ≤

0.01) and 19.76 (p ≤ 0.01) fold following high and low dose LPS treatment, respectively

(Figure 1H). Furthermore, at 6 h.p.i., the low dose LPS treated group was not statistically

different from the PBS control group, whereas the high dose treated group remained up-

regulated with a 1.92 fold increase (p ≤ 0.05). However by 24 h.p.i., iNOS transcript

levels in both the high and low dose treated groups were significantly down regulated by

4.07(p ≤ 0.05) and 3.45 (p ≤ 0.01) fold, respectively. Additionally, MHC-II transcripts

were increased by 1.80 fold (p ≤ 0.01) at 2 h.p.i. for the high dose treated group (Figure

1I). By 6 h.p.i., expression levels in both groups were not statistically different from the

control group. However, at 24 h.p.i transcripts in the high dose treated group were

reduced by 3.12 fold (p ≤ 0.01).

Expression of cytokine transcripts in the spleen following CpG and non-CpG ODN

treatment

The changes in gene expression levels among chickens treated with low dose (10

μg) or high dose (50 μg) of CpG ODNs, in addition to non-CpG ODNs (50 μg) are

expressed as fold changes relative to the PBS treated control group (Figure 2). For all

Page 59: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

45

genes examined, there were generally no statistically significant differences between the

non-CpG treated group and the PBS treated control group.

With respect to the type I interferon, IFN-α, we observed no significant

differences of IFN-α transcript levels among any of the treatment groups at any time

point (Figure 2A).

In contrast to IFN-α, we observed significant differences in transcripts of the

cytokines associated with the modulation and regulation of the immune response, namely

IFN-γ, IL-13 and IL-10. IFN-γ transcripts were up-regulated by 1.91 fold (p ≤ 0.01) for

the low dose treated group (Figure 2B). At 6 h.p.i., transcript levels in all groups were not

statistically different from the PBS control group. Moreover, IL-13 transcripts were

down-regulated at 2 h.p.i. for the high dose treated group by 1.93 fold (p ≤ 0.05) (Figure

2C). While transcript levels for the high dose treated group were not statistically different

from the PBS treated control group at 6 h.p.i., the low dose treated group had a 4.39 fold

(p ≤ 0.01) decrease, and remained decreased at 12 h.p.i. Nonetheless, at 24 h.p.i., both the

high and low dose CpG treated groups showed significant down-regulation in IL-13

transcripts by 1.96 (p ≤ 0.05) and 1.57 (p ≤ 0.05) fold, respectively. Furthermore, IL-10

transcripts were up-regulated at 2 h.p.i. by 2.37 fold (p ≤ 0.05) for the high dose treated

group (Figure 2D). However, at 6 h.p.i., transcript levels were not statistically different

from the control group, while transcript levels in the low dose CpG treated group

decreased by 2.6 fold (p ≤ 0.01). At 12 h.p.i., transcript levels in all treatment groups

were not statistically different from the PBS control group, but at 24 h.p.i., IL-10

transcripts were up-regulated for the high dose treated group by 1.61 fold (p ≤ 0.05).

Page 60: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

46

Similar to LPS treatment, we observed an up-regulation in both of the studied pro-

inflammatory cytokines, IL-1β and IL-8. Interleukin-1β transcripts were significantly up-

regulated at 2 h.p.i., with a 1.82 fold (p ≤ 0.05) and 2.18 fold (p ≤ 0.05) increase for low

and high dose CpG treated groups, respectively (Figure 2E). However, at 6 h.p.i.,

transcript levels for the low dose treated group were not statistically different compared

to PBS treated controls, while the high dose treated group still had a 2.01 (p ≤ 0.01) fold

increase in transcripts. However, at 12 h.p.i., IL-1β transcript levels in all groups were not

statistically different from the PBS treated control group. With respect to IL-8, transcripts

were not up-regulated for any of the treatment groups until 12 h.p.i., where there was a

2.23 fold (p ≤ 0.05) increase for the low dose treated group (Figure 2F). This cytokine in

the high dose treated group was increased by 1.99 fold, but this was not statistically

significant. There was no statistically significant up-regulation of gene expression

following the 12 h.p.i. time point.

Lastly, we examined the transcriptional responses of relevant proteins and

transcription factors which are suggestive of TLR stimulation and APC activation.

MyD88 transcripts were significantly up-regulated by 2.0 fold (p ≤ 0.05) at 2 h.p.i. for the

high dose CpG treated group (Figure 2G). However at 6 h.p.i., transcript levels among all

treatment groups were not statistically different from the PBS treated control group.

Furthermore, at 6 h.p.i. iNOS transcripts were down-regulated by 1.8 fold (p ≤ 0.01) and

by 3.05 fold (p ≤ 0.01) for the low dose treated group and the non-CpG treated group,

respectively (Figure 2H). However, at 12 h.p.i., iNOS transcripts were increased by 1.66

(p ≤ 0.05) fold for the low dose treated group. At 24 h.p.i., transcript levels were

decreased by 3.05 (p ≤ 0.01) and 2.5 (p ≤ 0.01) fold for low and high dose treated groups,

Page 61: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

47

respectively. Finally, MHC-II transcripts were not statistically different from the PBS

control group at all time points, except at 6 h.p.i., where the high dose treated group had a

16.94 fold (p ≤ 0.01) increase (Figure 2I).

A summary of all transcriptional responses of the examined genes in response to

LPS and CpG ODN treatments is presented in Table 4.

Page 62: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

48

Table 4. Summary of transcriptional responses in the spleen following LPS and CpG

ODN treatment.

Treatment Hours IFN-α IFN-γ IL-13 IL-10 IL-1β IL-8 MyD88 iNOS MHC II

LPS

100 μg

2 0 +++ 0 +++ +++ +++ + ++ 0

6 0 0 - + + + 0 0 0

12 0 0 0 0 + + 0 - 0

24 0 0 - 0 0 0 0 - 0

LPS

500 μg

2 + +++ + +++ +++ +++ + ++ +

6 0 + - ++ + + 0 + 0

12 0 0 - + + + 0 0 0

24 0 0 - - + + 0 - -

CpG

10 μg

2 0 + 0 0 + 0 0 0 0

6 0 0 - - 0 0 0 - 0

12 0 0 - 0 0 + 0 + 0

24 0 0 - 0 0 0 0 - 0

CpG

50 μg

2 0 0 - + + 0 + 0 0

6 0 0 0 0 + 0 0 0 ++

12 0 0 0 0 0 0 0 0 0

24 0 0 - + 0 0 0 - 0

A "+++" indicates a significant up-regulation of at least 20 fold, "++" indicates a

significant up-regulation of at least 5-fold, while a “+” indicates a significant up-

regulation when compared to the PBS treated control group. Furthermore, a “-” indicates

a significant down-regulation and a “0” indicates no significant change observed when

compared to the PBS treated control group.

Page 63: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

49

Discussion

Recognition of PAMPS by TLRs triggers innate host responses, promoting

secretion of cytokines, which leads to elicitation of innate and adaptive immune

responses (Medzhitov, 2001). In chickens, TLR agonists have been successfully used as

vaccine adjuvants (Linghua et al., 2007; Wang et al., 2009; Mallick et al., 2011) and as

prophylactic agents to prevent infection or ameliorate disease (Gomis et al., 2004; Dar et

al., 2009b). However, little is known about the dynamics and repertoire of chicken

responses to TLR agonists. Therefore, the present study was aimed to determine the

effects of stimulation with TLR4 and TLR21 ligands in chickens, by assessing

transcriptional regulation of a few relevant immune system genes in vivo.

Based on our findings, it appears that LPS elicits an overall greater response in

comparison to CpG ODNs. This could be a result of the shorter in vivo half-life of nucleic

acids as they are more prone to extracellular degradation. Moreover, similar to

mammalian TLR9, TLR21 may be expressed primarily in intracellular vesicles.

Therefore, it is possible that only a small fraction of the administered CpG ODN was

endocytosed and reached TLR21, therefore reducing the frequency of CpG-TLR21

interactions. To increase the intracellular availability of CpG ODNs, future studies should

consider incorporating CpG ODN into a protective vehicle, such as liposomes, to prevent

extracellular degradation and promote efficient delivery of CpG ODNs into the

endosome. Furthermore, our results indicate a temporal difference in peak responses to

CpG ODN and LPS. Following treatment with LPS, responses typically peaked by the

earliest time point, 2 hours post-treatment, while responses to CpG ODN peaked at

varying time points, both early and late. These findings agree with previously published

Page 64: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

50

time-series reports comparing the responses to CpG ODN and LPS in mammals

(Hartmann and Krieg, 1999). The increased response time to CpG ODN treatments may

again be related to the time required for uptake and interiorization of CpG ODNs by host

cells.

In mammals, TLRs are expressed by a variety of cell types including those of the

innate and adaptive immune system such as macrophages, dendritic cells (DCs), natural

killer (NK) cells, mast cells, T cells and B cells. Furthermore the response of these cells

to TLR stimulation has been well characterized. For example, treatment with TLR

agonists induces the proliferation of murine splenocytes and human peripheral blood

mononuclear cells, in addition to the up-regulation of cytokines, including type I IFNs,

pro-inflammatory cytokines as well as cytokines associated with TH1, TH2 and T

regulatory cells (Weeratna et al., 2005; Ghosh et al., 2007). Macrophages and DCs

express a wide array of TLRs in mammals and, therefore, play an important role in the

overall host response to TLR agonists. Indeed, it has been shown that TLR-activated

macrophages and DCs up-regulate expression of MyD88, co-stimulatory molecules and

MHC-II, in addition to up-regulating pro-inflammatory cytokines such as IL-1β and IL-8

(Ucla et al., 1990). These activated macrophages also increase their expression of iNOS,

leading to production of nitric oxide (Mosser, 2003). These findings may also be

extended to chickens, as previous studies have demonstrated production of nitric oxide

and pro-inflammatory cytokines by chicken heterophils and monocytes following

stimulation with TLR ligands (Kogut et al., 2005; He, MacKinnon, et al., 2011).

IFN-α and IFN-β belong to a class of antiviral cytokines that promote an antiviral

state through the production of interferon-inducible genes (Samuel, 2001). Typically,

Page 65: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

51

IFN-α is up-regulated during the early phases of a viral infection by virus-infected cells

and by TLR stimulated plasmacytoid dendritic cells (Doly et al., 1998). In addition to

their direct anti-viral effects, type I interferons promote the differentiation of naïve CD4+

T cells into TH1 cells (Brinkmann et al., 1993), enhancing their effector functions against

viruses. In the case of LPS, there was an early up-regulation of IFN-, raising the

possibility that LPS may directly and indirectly activate anti-viral responses through IFN-

γ and IFN-.

Regulation of an immune response is achieved through a variety of molecular

and cellular mechanisms, among which regulatory cytokines such as TGF-β and IL-10

are of note. IL-10 has anti-inflammatory properties which down-regulate activated

dendritic cells and macrophages, in addition to suppressing both TH1 and TH2 responses

(Saraiva and O’Garra, 2010). The results of the present study suggest that LPS up-

regulates IL-10 immediately following treatment. Importantly, LPS stimulation induced a

significant increase (50-100 fold increase depending on LPS dose) in IL-10 transcripts. It

is speculated that this immediate up-regulation of IL-10 may serve as a protective

mechanism to regulate the activation of responding cells. It is not clear which cells were

the predominant source of IL-10, however, it is plausible that macrophages, DCs, T cells

and B cells, were the main producers of this cytokine, as they have high expression of

TLR4 and can respond to TLR agonists by producing IL-10 (Barr et al., 2007; Conroy et

al., 2008).

Although LPS is common to all Gram negative bacteria, kinetics of host response

to LPS is partly dependent on the bacterial species the LPS is derived from. As

demonstrated by Keestra and colleagues (2008), chicken TLR4 responds more robustly to

Page 66: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

52

certain LPS including those derived from E. coli and Salmonella spp as determined by a

NF-κappaβ reporter assay. Furthermore in mammals, treatment with LPS derived from

Salmonella spp and Escherichia coli promotes a TH1 biased response characterized by the

up-regulation of IL-12 and IFN-γ, while treatment with LPS derived from

Porphyromonas gingivalis does not up-regulate either of these two cytokines, but

promotes the production of IL-5, which is suggestive of a TH2-like response (Pulendran

et al., 2001). Although responses to multiple sources of LPS were not characterized in the

present study, our results indicate that LPS derived from E. coli up-regulates both IFN-γ

and IL-13 in the spleen, raising the possibility of a more balanced TH1/ TH2-like

phenotype, although further studies are needed to confirm this. Furthermore, IL-13 may

synergize with IL-10 to promote the resolution of the immune response, as previous

studies have demonstrated the anti-inflammatory properties of IL-13 (Marie et al., 1996;

Watson et al., 1999).

A similar ligand specific response applies to TLR21 as well, as the sequences

flanking the CpG motif differentially modulate the kinetics of the host response. There

are three major classes of CpG ODN that are grouped according to their flanking

sequence motifs, classes A, B and C (Vollmer et al., 2004). The class A CpG ODNs are

potent activators of mammalian TLR9 expressed by plasmacytoid dendritic cells (pDC),

thereby promoting the production of IFN-α (Krug et al., 2001). This is in contrast to class

B CpG ODN which weakly induces IFN-α. However, this ODN is strongly

immunostimulatory for B cells, thereby enhancing proliferation and antigen presentation

capabilities, in addition to the promoting the production of cytokines such as IFN-γ (Krug

et al., 2001). Class C CpG ODN contains a mixture of Class A and Class B flanking

Page 67: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

53

sequences, thereby moderately activating both pDCs and B cells. Although the responses

to different classes of CpG ODN in chickens remain to be elucidated, it has been shown

that chicken B cells proliferate in response to Class B and Class C CpG ODNs (Wattrang,

2009), which may support the notion of a flanking-sequence specific response

demonstrated by chicken TLR21. In the present study, we administered the class B CpG

ODN 2007 and subsequently observed no up-regulation of IFN-α in the spleen, which is

similar to IFN-α-independent responses that occur in mammals in response to class B

CpG ODN (Liu et al., 2010). Furthermore, we observed a significant up-regulation of

IFN-γ and MHC II in the spleen, which may be attributed potentially to a robust TLR-

mediated B cell response (Jiang et al., 2007; Booth et al., 2010). The biological

significance of IFN-γ has been well established in mammals, such as promoting the

activation of cell subsets with anti-viral capacity including cytotoxic T lymphocytes,

macrophages and natural killer cells (Samuel, 2001). Additionally, IFN-γ possesses anti-

viral properties as well, inducing virus-infected cells to produce and activate anti-viral

proteins such as RNase L, which inhibit viral replication and promote apoptosis of the

infected cell (Samuel, 2001). Furthermore, IFN-γ is indicative of a TH1-biased response,

and therefore our data raise the possibility that treatment with Class B CpG ODN biases

the immune response towards a TH1 phenotype in chickens, as characterized by the up-

regulation of IFN-γ and down-regulation of IL-13, although further studies are needed to

confirm this.

Lastly, we observed an up-regulation of IL-10 in response to CpG ODN

treatment, likely functioning to promote resolution of the immune responses and regulate

the activation of responding cells. These findings support previous in vitro studies, which

Page 68: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

54

suggested that dendritic cells and B cells produce IL-10 in response to CpG ODN (Lenert

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

In conclusion, the present study highlights some of the underlying mechanisms of

induction of host responses by TLR agonists in chickens. Future studies may employ

these findings to select TLR ligands for enhancing immune responses in chickens, for

example for enhancing immunity conferred by vaccines.

Page 69: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

55

IFN-

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0

1

2

3

4

55

10

15

LPS - 100 g

LPS - 500 g*

IFN-

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0

1

2

3

4150

200

250

300

350

400

LPS - 100 g

LPS - 500 g

**

**

*

IL-13

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4LPS - 100 g

LPS - 500 g

*

** *** **

IL-10

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0

1

2

3

5

10

15

50

100

150LPS - 100 g

LPS - 500 g*

**

**

**

*

**

IL-1

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

2

4

6

8

10

50

100

150

200

LPS - 100 g

LPS - 500 g**

**

** ** ***

*

IL-8

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0

2

4

6

8

10

20

30

40

50 LPS - 100 g

LPS - 500 g** **

*

** *

**

**

MyD88

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

2

4

6

8

10LPS - 100 g

LPS - 500 g*

*

iNOS

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0

1

2

3

410

20

30

40

50

LPS - 100 g

LPS - 500 g** **

*

* ** *

MHC II

Time P.I.

Fo

ld

Ch

an

ge

2h 6h 12h

24h

0.0

0.5

1.0

1.5

2.0

2.5

LPS - 100 g

LPS - 500 g**

**

A CB

D FE

G IH

Figure 1. Gene expression in the spleens of chickens at 2, 6, 12 and 24 hours post

injection (h.p.i.) with 100 μg or 500 μg of LPS. Gene expression was assessed using

quantitative RT-PCR, and was calculated relative to the house keeping gene β-actin.

Graphed values are expressed as fold change relative to the PBS treated control group ±

standard error. Results were considered statistically significant from the PBS treated

control group if p ≤ 0.05 (*) and p ≤ 0.01 (**).

Page 70: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

56

IFN-

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4

5 CpG - 10 g

CpG - 50 g

Non CpG - 50 g

IFN-

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4

5 CpG - 10 g

CpG - 50 g

**

Non CpG - 50 g

IL-13

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0.0

0.5

1.0

1.5

2.0

CpG - 10 g

CpG - 50 g

Non CpG - 50 g

*

**

*

**

IL-10

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4 CpG - 10 g

CpG - 50 g

* Non CpG - 50 g

**

*

IL-1

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4CpG - 10 g

CpG - 50 g

Non CpG - 50 g*

***

IL-8

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4

5 CpG - 10 g

CpG - 50 g

Non CpG - 50 g

*

MyD88

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

4CpG - 10 g

CpG - 50 g

Non CpG - 50 g*

iNOS

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3 CpG - 10 g

CpG - 50 g

Non CpG - 50 g

****

*

** **

MHC II

Time P.I.

Fo

ld C

han

ge

2h 6h 12h

24h

0

1

2

3

5

10

15

20 CpG - 10 g

CpG - 50 g**

Non CpG - 50 g

A CB

D FE

G IH

Figure 2. Gene expression in the spleens of chickens at 2, 6, 12 and 24 hours post-

injection (h.p.i.) with 10 μg or 50 μg of CpG ODNs, or 50 μg of non CpG ODNs . Gene

expression was assessed using quantitative RT-PCR, and was calculated relative to the

house keeping gene β-actin. Graphed values are expressed as fold change relative to the

PBS treated control group ± standard error. Results were considered statistically

significant from the PBS treated control group if p ≤ 0.05 (*) and p ≤ 0.01 (**).

Page 71: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

57

CHAPTER 3

Prophylactic treatment with Toll-like receptor ligands enhances host immunity to

avian influenza virus in chickens

Michael St. Paul a, Amirul I. Mallick

ab, Leah R. Read

a, Alexander Ian Villanueva

a,

Payvand Parvizi a, Mohamed Faizal Abdul-Careem

c Éva Nagy

a and

Shayan Sharif

a*.

a Department of Pathobiology, Ontario Veterinary College, University of Guelph,

Guelph, Ontario, Canada, N1G 2W1.

b Present address: Department of Imaging Research, Sunnybrook Research Institute,

Sunnybrook Health Science Centre, University of Toronto, 2075 Bayview Avenue,

University of Toronto, Toronto, Ontario, Canada, M4N 3M5.

c Department of Ecosystem and Public Health Faculty of Veterinary Medicine, University

of Calgary, Calgary, Alberta, Canada, T2N 4N1.

*To whom correspondence should be addressed:

Dr. Shayan Sharif, Department of Pathobiology, Ontario Veterinary College, University

of Guelph, Guelph, Ontario, N1G 2W1, Canada.

Tel: 1 (519) 824-4120 ext. 54641, Fax: 1(519) 824-5930 Email: [email protected]

Published in Vaccine 2012; 30(30): 4524-31

Page 72: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

58

Abstract

Avian influenza viruses (AIV) pose a threat towards the health of both poultry

and humans. To interrupt the transmission of the virus, novel prophylactic strategies must

be considered which may reduce the shedding of AIV. One potential is the prophylactic

use of Toll-like receptor (TLR) ligands. Many cells of the immune system express TLRs,

and cellular responses to TLR stimulation include activation and the production of

cytokines. TLR ligands have been employed as prophylactic treatments to enhance host

resistance to pathogens both in mammals and chickens. Therefore, the present study was

conducted to determine whether TLR ligands may be used prophylactically in chickens to

enhance host immunity to AIV. Chickens received intramuscular injections of either low

or high doses of the TLR ligands poly I:C, lipopolysaccharide (LPS) and CpG ODN.

Twenty-four hours post-treatment, chickens were infected with the low pathogenic avian

influenza virus H4N6, and both oropharyngeal and cloacal virus shedding were assessed

on days 4 and 7 post-infection. To identify potential correlates of immunity, spleen and

lungs were collected on days 2, 4 and 7 post-infection for RNA extraction. The results

suggested that all of the TLR ligand treatments induced a significant reduction in virus

shedding, with the TLR3 ligand poly I:C conferring the greatest AIV immunity compared

to control birds, followed by CpG ODN and LPS. Furthermore, transcriptional analysis of

gene expression in the spleen and lungs suggest IFN-α and IL-8 as correlates of immunity

conferred by poly I:C, and IFN-γ for CpG ODN and LPS. In conclusion, TLR ligands,

have the ability to enhance host immunity against AIV, and future studies should

consider exploring the combinatory effects of poly I:C and CpG ODN prophylaxis in

conjunction with AIV vaccination.

Page 73: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

59

1. Introduction

Avian influenza viruses (AIV) are enveloped, single stranded RNA viruses

belonging to the family Orthomyxoviridae. AIV are typically classified according to their

pathogenicity; whether they are a low pathogenic avian influenza (LPAI) virus or a

highly pathogenic avian influenza virus (HPAI) (Suarez and Schultz-Cherry, 2000).

Although HPAI viruses, such as H5N1, are of great concern to the poultry industry and

public health, outbreaks of LPAI viruses have also become problematic. For example, the

recent spread of H7N3 among poultry in England led to the culling of 35,000 chickens

(Nguyen-Van-Tam et al., 2006), while an outbreak of H5N2 in Japan affected 5.78

million chickens spread across 41 farms (Okamatsu et al., 2007). Importantly, some LPAI

strains such as H9N2, have the ability to transmit to other species including to humans

and pigs (Lin et al., 2000). To interrupt the inter- and intra-species transmission of AIV

from infected poultry, several prophylactic strategies may be considered, including

vaccination, administration of antiviral compounds such as interferons, as well as the use

of Toll-like receptor ligands.

Toll-like receptors (TLRs) are an evolutionarily conserved group of pattern

recognition receptors that play an important role in the initiation and modulation of host

responses to pathogens. Ten TLRs have been identified in chickens, each recognizing

conserved structural motifs of pathogens termed pathogen-associated molecular patterns

(PAMPs) (Temperley et al., 2008). For example, TLR3 recognizes double stranded RNA,

a product of some viruses during their replication cycle, as well as synthetic molecules

such as such as polyinosinic:polycytidylic acid (poly I:C), while TLR4 recognizes

lipopolysaccharide (LPS) found in the outer membrane of Gram-negative bacteria, and

Page 74: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

60

TLR21, the functional homologue of mammalian TLR9, recognizes CpG DNA motifs

found in certain bacterial and viral nucleic acids (Medzhitov, 2001; Brownlie et al.,

2009). TLRs are expressed by a variety of immune system cell subsets where they

mediate cellular activation and the production of cytokines (Hopkins and Sriskandan,

2005; Akira et al., 2006).

TLR ligands have been employed prophylactically in laboratory animal models to

enhance host resistance to pathogens including hepatitis C virus, Burkholderia mallei and

influenza virus (Waag et al., 2006; Kanzler et al., 2007; Wong et al., 2009). Moreover,

TLR ligand prophylaxis has been extended to chickens, enhancing host immunity to

pathogens including Salmonella enterica serovar Enteritidis (Genovese et al., 2007; He et

al., 2007b), Escherichia coli (Gomis et al., 2004; Taghavi et al., 2009), Marek’s disease

virus (P. Parvizi, unpublished data) and infectious bronchitis virus (Dar et al., 2009b).

Therefore, we hypothesized that TLR ligands may enhance immunity to AIV as well,

thereby reducing viral load and shedding from chickens that are experimentally infected

with AIV. The results of our study suggest that the ligands for TLR3, 4 and 21 can reduce

virus load and shedding in infected birds, with the TLR3 ligand poly I:C conferring the

greatest host immunity towards AIV.

Page 75: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

61

2. Materials and Methods

2.1 Experimental chickens and housing

Specific pathogen-free (SPF) eggs (n=108) were procured from the Animal Disease

Research Institute, Canadian Food Inspection Agency (Ottawa, ON), hatched at the

Arkell Poultry Research Center, University of Guelph (Guelph, ON) and housed in the

isolation facility of the Ontario Veterinary College, University of Guelph. This research

was approved by the University of Guelph Animal Care Committee and complied with

the guidelines of the Canadian Council on Animal Care.

2.2 Avian influenza virus

Chickens were infected with the LPAI A/Duck/Czech/56 (H4N6) which was propagated

in 11-day-old embryonated chicken eggs as described in (Mallick et al., 2011).

2.3 TLR ligands

Poly I:C and LPS from E. coli 0111:B4 were purchased from Sigma-Aldrich, Canada

(Oakville, ON), while synthetic class B CpG ODN 2007 [5’-

TCGTCGTTGTCGTTTTGTCGTT-3’] and non-CpG ODN [5’-

TGCTGCTTGTGCTTTTGTGCTT-3’] were purchased from Eurofins MWG Operon

(Ebersberg, GER). All of the ligands used were re-suspended in sterile phosphate

buffered saline (PBS, pH 7.4).

Page 76: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

62

2.4 Experimental design

Fourteen-day-old chickens were randomly divided into 9 groups of 12 birds each.

Chickens were injected intramuscularly (i.m.) in the pectoral muscle with either a low (80

μg) or high (400 μg) dose of poly I:C, a low (100 μg) or high (500 μg) dose of LPS or a

low (10 μg) or high (50 μg) dose of CpG ODN, while control groups received either

non-CpG ODN (50 μg) or PBS (St Paul et al., 2011; Parvizi et al., 2012). Twenty-four

hours post-treatment (Renis, 1970; Ashkar et al., 2003; Dar et al., 2009b), in all of the

groups except for one of the two PBS treated control groups, chickens were infected with

1x106

50% tissue culture infectious dose (TCID50) of H4N6 virus through the oculo-nasal

route. To assess virus shedding on days 4 and 7 post-infection (p.i.), oropharyngeal and

cloacal swabs were collected from 4 to 6 chickens in each group, depending on the

number of chickens remaining. These time points represent the peak in oropharyngeal

and cloacal shedding, respectively (Le Gall-Reculé et al., 2007; Spackman et al., 2010).

To identify correlates of immunity, 4 chickens from each group were euthanized on days

2, 4 and 7 p.i., and spleen and lung tissue were stored in RNA later (Qiagen, Valencia,

CA). Of the two doses of TLR ligands administered to the chickens, only mRNA

expression in the spleen and lung tissue of the most efficacious dose of each TLR agonist

(poly I:C high dose, LPS low dose and CpG ODN high dose) was quantified.

2.5 Virus titration

Oropharyngeal and cloacal swabs were collected as described (WHO, 2002). A virus

titration assay using Madin-Darby canine kidney (MDCK) cells (University of Guelph

Page 77: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

63

Animal Health Laboratory) was performed to assess viral shedding from both

oropharyngeal and cloacal swabs as described previously (Szretter et al., 2006). Viral

titres were expressed as log10 TCID50/mL of swab sample.

2.6 RNA extraction, cDNA synthesis and real-time PCR

Total RNA extraction and cDNA synthesis was performed as previously described (St

Paul et al., 2011). Real-time PCR using SYBR Green was performed on diluted cDNA

using the LightCycler® 480 II (Roche Diagnostics GmbH, Mannheim, GER) as

previously described (Villanueva et al., 2011). Primers were synthesized by Sigma-

Aldrich, Canada (Oakville, ON), and their specific sequences have been previously

published elsewhere (Abdul-Careem et al., 2006, 2008a, 2009; Brisbin et al., 2008;

Villanueva et al., 2011).

2.7 Data analysis

The log10TCID50/mL of virus present in the oropharyngeal and cloacal swab fluid of each

chicken was calculated based on the formula by Reed-Muench (Szretter et al., 2006). To

examine if each TLR ligand, both high and low doses, differed from the PBS control,

separate ANOVA for each TLR ligand were computed. Any statistically significant main

effects were examined with post-hoc Tukey’s test. With respect to quantitative real-time

PCR, relative expression of all target genes was calculated relative to the housekeeping

gene β-actin using the LightCycler® 480 Software (Roche Diagnostics GmbH,

Page 78: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

64

Mannheim, GER), based on the formula developed by Pfaffl (Pfaffl, 2001). Statistical

difference was calculated using an un-paired student’s t-test. All results were considered

statistically significant from the infected, PBS-treated control group if p ≤ 0.05 (*) and p

≤ 0.01 (**), considered statistically significant from the uninfected PBS control group if

p ≤ 0.05 (#), and lastly in the case of CpG ODN, considered statistically significant from

the non-CpG ODN group if p ≤ 0.05 (~).

Page 79: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

65

3. Results

3.1 Treatment with TLR ligands reduces oropharyngeal and cloacal virus shedding.

On days 4 and 7 p.i., oropharyngeal and cloacal virus shedding was assessed by

virus titration in MDCK cells (Figure 3). Treatment with the high dose of poly I:C

resulted in the lowest amount of virus being shed orally on day 4 p.i. (Figure 3A), with a

log10 TCID50/mL of 1.84 (p ≤ 0.01), followed by, from lowest to highest, treatment with

the low dose of poly I:C (2.92 log10 TCID50/mL, p ≤ 0.01), the high dose of CpG ODN

(3.17 log10 TCID50/mL, p ≤ 0.01), the low dose of LPS (4.59 log10 TCID50/mL, p ≤ 0.01),

and lastly, treatment with the low dose of CpG ODN (5.50 log10 TCID50/mL, p ≤ 0.01).

Of the two control groups which were infected, the non-CpG ODN treated group had a

viral titre of 4.59 log10 TCID50/mL (p ≤ 0.01), while the PBS treated group had a viral

titre of 6.92 log10 TCID50/mL. Although a similar pattern was observed for cloacal

shedding on day 4 p.i. (Figure 3B), virus shedding in none of the treatment groups

resulted in a statistically significant difference from the PBS treated control group.

There was no detectable oropharyngeal shedding on day 7 p.i. in any of the

treatment and control groups (data not shown). However, shedding was detected in the

cloaca on day 7 p.i., and treatment with the high dose of CpG ODN resulted in the lowest

amount of virus being shed (Figure 3C), with a titre of 1.75 log10TCID50/mL (p≤ 0.01),

followed by, from lowest to highest, treatment with the high dose of poly I:C (2.17

log10TCID50/mL, p≤ 0.01), the low dose of poly I:C (2.75 log10TCID50/mL, p≤ 0.01), the

low dose of LPS (3.09 log10TCID50/mL, p≤ 0.05) and lastly, treatment with the low dose

of CpG ODN (3.25 log10TCID50/mL, p≤ 0.01). Of the two control groups that were

Page 80: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

66

infected, the non-CpG ODN treated group had a viral titre of 3.25 log10 TCID50/mL (p ≤

0.01), while the PBS treated group had a viral titre of 5.92 log10 TCID50/mL.

3.2 Correlates of immunity in the spleen and lungs of chickens treated with TLR

ligands.

To identify immunological correlates of protection, gene expression in the spleen

and lungs was examined on days 2, 4 and 7 p.i. using real-time PCR (Figures 4-7).

Statistical significance described in the results is in reference to the infected, PBS treated

control group unless otherwise indicated.

Interferon (IFN)-α transcripts were down-regulated in the spleen on day 7 p.i. in

the CpG ODN treated group (p≤ 0.01) (Figure 4A), while IFN-β transcripts were up-

regulated in the LPS treated group on day 4 p.i. (p≤ 0.01) (Figure 4B). With respect to

IFN-γ, transcripts were down-regulated in the spleens of the poly I:C, LPS, CpG and non-

CpG ODN treated groups on day 7 p.i. (p≤ 0.01) (Figure 4C). Transcripts of the

interferon inducible gene 2’-5’-oligoadenylate synthetase (OAS) were up-regulated (p≤

0.05) in the CpG ODN treated group in the spleen on day 4 p.i., (Figure 4D).

In the spleen on day 2 p.i., transcript levels of IL-1β were down-regulated in the

CpG ODN treated group (p≤ 0.05) (Figure 5A), As for IL-8, there were no statistically

significant differences in transcript levels for any of the treatment groups in the spleen

(Figure 5B). Transcript levels of IL-18 were significantly down-regulated in the spleen

on day 7 p.i. in the poly I:C treated group (p≤ 0.05), and in the LPS, CpG ODN and non-

CpG ODN treated groups (p≤ 0.01) (Figure 5C).

Page 81: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

67

In the lungs, IFN-α transcripts were up-regulated in the poly I:C treated group on

day 4 p.i. (p≤ 0.05) and day 7 p.i. (p≤ 0.05), and up-regulated in the LPS treated group on

day 7 p.i. as well (p≤ 0.01) (Figure 6A). Similarly, IFN-β transcripts were up-regulated in

the lungs on day 2 p.i. in the LPS, poly I:C, CpG ODN and non-CpG ODN treated groups

(p≤ 0.05) (Figure 6B). However, on day 4 p.i., transcripts were down-regulated in the

poly I:C and non-CpG ODN treated groups (p≤ 0.05). In the case of IFN-γ, transcripts

were up-regulated in the lungs on day 2 and 4 p.i. for the CpG ODN treated group (p≤

0.01), while being up-regulated only on day 4 p.i. for the LPS and non-CpG ODN treated

groups (p≤ 0.05) (Figure 6C). On day 7 p.i., IFN-γ transcripts in the poly I:C treated

group were up-regulated (p≤ 0.05). Transcripts for OAS were up-regulated in the lungs

on day 4 p.i., in the CpG ODN treated group (p≤ 0.05) (Figure 6D).

In the lung, transcripts for IL-1β were up-regulated in the CpG ODN treated

group on day 7 p.i. (p≤ 0.05) (Figure 7A), while IL-8 transcripts were up-regulated on

day 2 p.i. (p≤ 0.01) in the poly I:C treated group (Figure 7B). Lastly, IL-18 transcripts

were up-regulated in the lungs on day 2 p.i. in the poly I:C treated group (p≤ 0.05), while

being down-regulated in the LPS, CpG ODN and non-CpG ODN treated groups on day 7

p.i. (p≤ 0.05) (Figure 7C). A summary of virus shedding and mRNA expression is found

in Table 5.

Page 82: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

68

Table 5. Summary of virus shedding and transcript levels in the lungs on days 4 and

7 p.i.

TLR

ligand

Day

P.i.

Reduction in

Oropharyngeal

Shedding

Reduction

in Cloacal

Shedding

IFN-α IFN-β IFN-γ OAS IL-1β IL-8 IL-18

Poly

I:C

(high

dose)

4 5.08 0.96 + - 0 0 0 0 0

7 ND 3.75 + 0 + 0 0 0 0

LPS

(low

dose)

4 2.33 0.33 0 0 + 0 0 0 0

7 ND 2.83 ++ 0 0 0 0 0 -

CpG

ODN

(high

dose)

4 3.75 0.71 0 0 ++ + 0 0 0

7 ND 4.17 0 0 0 0 + 0 -

Non-

CpG

ODN

4 2.33 0.12 0 - + 0 0 0 0

7 ND 2.67 0 0 0 0 0 0 -

The amount reduction in virus shedding was determined by subtracting the mean log10

TCID50/mL for each treatment group from that of the infected, PBS treated control group.

A “ND” indicates no shedding was detected. A “+” and “++” indicate a significant up-

regulation of p ≤ 0.05 and p ≤ 0.01, respectively, when compared to the infected, PBS

treated control group. A “0” indicates no significant up-regulation and a “-“ indicates a

significant down-regulation compared to the infected, PBS treated control group.

Page 83: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

69

4. Discussion

The immunomodulatory properties of TLR ligands have been exploited to

enhance host immunity to several pathogens in chickens. In the present study, we report

that TLR ligands may also confer protection against an LPAI virus.

When used prophylactically, our findings indicate that all three of the TLR ligand

treatments reduced AIV shedding, with the TLR3 ligand poly I:C being the most

efficacious followed by CpG ODN and LPS. The anti-viral role of poly I:C has been well

established in mammals, especially in the context of enhancing influenza virus immunity

(Ichinohe et al., 2005; Wong et al., 2009). This anti-influenza property of poly I:C may

be attributed to the unique signaling pathway utilized by TLR3, which signals solely

through the TIR domain-containing adaptor inducing IFN- β (TRIF) pathway and thereby

promotes the production of the type I interferons IFN-α and IFN-β (Matsumoto and Seya,

2008). Consistent with these findings, we observed an up-regulation of type I IFNs in the

lungs of the poly I:C treated chickens, possibly mediated in part by TLR3 and other

pattern recognition receptors that also bind dsRNA, such as Mda5 (Karpala et al., 2011b).

Type I interferons demonstrate many anti-viral functions, including inhibiting viral

transcription and translation, promoting the apoptosis of infected cells and activating

antigen presenting cells (Samuel, 2001). Furthermore, IFN-α has been shown to protect

ferrets (Kugel et al., 2009) and chickens (Meng et al., 2011) against influenza viruses,

and may also contribute to viral clearance during the natural course of infection, as our

data indicate an up-regulation of IFN- α in the spleens of the challenged control group.

In addition to IFN- α, our results suggest that poly I:C significantly up-regulated IL-8 in

the lungs, raising the possibility that heterophils were recruited to the lung, due to the fact

Page 84: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

70

that IL-8 is a chemoattractant for heterohpils (Kogut, 2002). Although the importance of

heterophils in AIV immunity is not clear, in mammals neutrophils promote the clearance

of influenza virus in part through the production of cytokines and anti-microbial peptides

(Gaudreault and Gosselin, 2008; Wang et al., 2008). Taken together, our results suggest

that IFN-α and IL-8 both serve as correlates of AIV immunity for the poly I:C treatment.

In chickens, CpG ODN has previously been shown to induce a TH1 biased

immune response characterized by the up-regulation of IFN-γ and down-regulation of IL-

13 (Patel et al., 2008; St Paul et al., 2011). Although not as efficacious as poly I:C, our

results showed that treatment with both low and high doses of CpG ODN significantly

reduced AIV shedding. Moreover, our data suggest that immunity conferred by CpG

ODN correlates with IFN-γ, which promotes an anti-viral state mediated in part by CD8+

T cells and natural killer (NK) cells. Furthermore, IFN-γ induced the up-regulation of the

interferon inducible gene 2’-5’ OAS thereby suggesting the initiation of the RNase L

pathway, which acts to cleave viral RNA and promote the apoptosis of infected cells

(Liang et al., 2006). Interestingly, our results suggest that treatment with non-CpG ODN

enhanced immunity to AIV as well, although it was not as efficacious as an equivalent

dose of CpG ODN. Similar immunostimulatory effects of non-CpG ODN have been

observed both in mammals as well as in chicken B cells (Senn et al., 2005; Wattrang,

2009). Although our findings raise the possibility that the immunity conferred by non-

CpG ODN was mediated in part through the production of IFN-γ, further studies are

needed to elucidate the mechanisms in which non-CpG ODN modulate the chicken

immune system.

Page 85: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

71

In contrast to the other examined TLR ligands, LPS is strictly a bacterial PAMP.

Chickens treated with low dose LPS (but not high dose LPS) displayed reduced virus

shedding. Although the basis behind this observation is unknown, a similar phenomenon

has been observed in mice. Mice challenged with influenza virus PR8 strain and treated

with high doses of LPS (i.e. 20 μg/g) demonstrated an increased virus load which was

attributed to an influenza virus associated encephalopathy (Tanaka et al., 2010). This is in

contrast to mice treated with lower doses of LPS (i.e. 1.25 μg/g), as they are protected

against challenges with PR8 and the highly pathogenic H5N1 (Shinya et al., 2011). As

such, future studies may consider employing a lower range of LPS doses, or perhaps even

substituting LPS with another TLR4 ligand, such as a synthetic lipid A or FimH, which

have previously been shown to protect mice against influenza virus infection (Cluff et al.,

2005; Abdul-Careem et al., 2011). Our results suggest that type I interferons and IFN-γ

serve as potential correlates of immunity conferred by LPS, which is also supported by

our previous study which indicated an up-regulation of these cytokines in response to

LPS treatment (St Paul et al., 2011). In mice, it has previously been shown that immunity

to H5N1 conferred by LPS is dependent on signaling both through the myeloid

differentiation factor 88 (MyD88) independent pathway, as well as the presence of the

accessory molecules cluster of differentiation (CD)14 and myeloid differentiation

protein-2 (MD-2) (Shinya et al., 2012). In chickens, it has been suggested that responses

mediated by TLR4 is dependent in part on signaling through the MyD88 independent

pathway (Lian et al., 2012) as well relying on the presence of both CD14 and MD-2

(Kogut et al., 2005a; Keestra and van Putten, 2008). As such, we speculate that immunity

Page 86: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

72

to AIV conferred by LPS in chickens may be mediated by the same pathways and

accessory molecules identified in mice.

Through transcriptional analysis of relevant immune system genes, we discovered

that several of the genes were up-regulated in the lungs while being down-regulated in

the spleen. This finding might suggest that a greater proportion of the immune response

to AIV occurs in the lungs rather than in the spleen, which is in agreement with previous

studies (Degen et al., 2006; De Geus et al., 2011). Having said that, it is possible a

contributing factor towards the efficacy of an intramuscularly injected TLR ligand is its

ability to up-regulate immune system genes in the lungs. Previously, our group has

demonstrated that an i.m. injection of poly I:C in chickens induces a significant up-

regulation of many immune system genes in the lungs (Parvizi et al., 2012) however it

remains to be elucidated whether i.m. injections of LPS and CpG ODN up-regulate genes

in the lungs as well, and if so, to what extent when compared to that of poly I:C.

Although TLR ligand prophylaxis may be efficacious in an experimental setting,

there are some limitations that need to be addressed prior to their widespread adoption.

For example, it has previously been shown in chickens that the protective benefits

conferred by the ligands is highest when the challenge occurs within 3 days of treatment,

and decreases if this time period is extended to 6 days and is absent at 9 days and

onwards (Gomis et al., 2003). Therefore, we propose that future research should be aimed

at investigating ways to increase the duration of efficacy of TLR ligands while at the

same time facilitating their administration. One potential strategy may be to protect the

TLR ligands from degradation by encapsulating them in protective vehicles, such as

nanoparticles, which can then be aerosolized (Evans et al., 2011),

Page 87: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

73

In conclusion, TLR ligands are able to reduce the viral load following infection with

an LPAI virus, with the TLR3 ligand poly I:C being the most efficacious in terms of

reducing the quantity of virus shed. Furthermore, transcriptional analysis of genes in the

spleen and lungs identified IL-8, IFN-α, IFN-β and IFN-γ as correlates of immunity.

Future studies should explore the prophylactic use of TLR agonists in conjunction with

vaccination against AIV, as this may potentially lead to a greater reduction in virus

shedding conferred by either treatment alone.

Page 88: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

74

Figure 3. Titre of AIV present in oropharyngeal and cloacal swabs on days 4 and 7 post-

infection. Twenty-four hours prior to infection, chickens received intramuscular

injections of poly I:C (80 μg or 400 μg), LPS (100 μg or 500 μg), CpG ODN (10 μg or 50

μg), non-CpG ODN (50 μg), or PBS. Viral titre was assessed by virus titration in MDCK

cells. Graphed values represent the mean log10TCID50/mL of swab fluid in each treatment

group, with error bars representing standard error. Statistical significance between

treatment groups compared to the infected, PBS treated control group was calculated

using an ANOVA followed with a post-hoc Tukey’s test, and results were considered

statistically significant if p ≤ 0.05 (*) and p ≤ 0.01 (**) and in the case of CpG ODN,

statistically significant from the non-CpG ODN group if p ≤ 0.05 (~).

Day 4 Cloacal

Treatment

log

10 T

CID

50

/mL

Poly

I:C L

ow

Poly

I:C H

igh

LPS L

ow

LPS H

igh

CpG

Low

CpG

Hig

h

Non-C

pGPBS

0

1

2

3

4

Day 4 Oropharyngeal

Treatment

log

10 T

CID

50

/mL

Poly

I:C L

ow

Poly

I:C H

igh

LPS L

ow

LPS H

igh

CpG

Low

CpG

Hig

h

Non-C

pGPBS

0

2

4

6

8

**

**

**

**

****

~

~

Day 7 Cloacal

Treatment

log

10 T

CID

50

/mL

Poly

I:C L

ow

Poly

I:C H

igh

LPS L

ow

LPS H

igh

CpG

Low

CpG

Hig

h

Non-C

pGPBS

0

2

4

6

8

** *** **

**

**

~

A B

C

Page 89: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

75

IFN- Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

2.010 - 9

4.010 - 9

6.010 - 9

**

#

#

#

#

IFN- Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

2.010 - 9

4.010 - 9

6.010 - 9

8.010 - 9

1.010 - 8

**

#

#

#

IFN- Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

1.010 - 9

2.010 - 9

3.010 - 9

4.010 - 9

5.010 - 9

## #

**

****

**##

OAS Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

2.010 - 9

4.010 - 9

6.010 - 9

8.010 - 9

#

#

*

~

DC

BA

Poly I:C LPS CpG Non-CpG PBS Unchallenged

Figure 4. Relative gene expression of interferons and the interferon inducible gene 2’-5’-

oligoadenylate synthetase (OAS) in the spleen on days 2, 4 and 7 post-infection as

determined by quantitative RT-PCR. Twenty-four hours prior to infection, chickens

received intramuscular injections of poly I:C (400 μg), LPS (100 μg), CpG ODN (50 μg),

non-CpG ODN (50 μg), or PBS. Graphed values represent mean gene expression levels

relative to the house keeping gene β-actin ± standard error. Statistical significance

between treatment groups compared to the infected, PBS treated control group was

calculated using an un-paired students t test, and results were considered statistically

significant if p ≤ 0.05 (*) and p ≤ 0.01 (**), considered statistically significant from the

uninfected PBS control group if p ≤ 0.05 (#), and in the case of CpG ODN, statistically

significant from the non-CpG ODN group if p ≤ 0.05 (~).

Page 90: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

76

Figure 5. Relative gene expression of the cytokines IL-1β, IL-8 and IL-18 in the spleen

on days 2, 4 and 7 post-infection as determined by quantitative RT-PCR. Twenty-four

hours prior to infection, chickens received intramuscular injections of poly I:C (400 μg),

LPS (100 μg), CpG ODN (50 μg), non-CpG ODN (50 μg), or PBS. Graphed values

represent mean gene expression levels relative to the house keeping gene β-actin ±

standard error. Statistical significance between treatment groups compared to the

infected, PBS treated control group was calculated using an un-paired students t test, and

results were considered statistically significant if p ≤ 0.05 (*) and p ≤ 0.01 (**),

considered statistically significant from the uninfected PBS control group if p ≤ 0.05 (#),

and in the case of CpG ODN, statistically significant from the non-CpG ODN group if p

≤ 0.05 (~).

IL-1 Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

5.010 - 1 0

1.010 - 0 9

1.510 - 0 9

2.010 - 0 9

2.510 - 0 9

*

#

#

#~

IL-8 Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

2.010 - 9

4.010 - 9

6.010 - 9

8.010 - 9

IL-18 Spleen

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

5.010 - 1 0

1.010 - 0 9

1.510 - 0 9

***** **

#

C

BA

Poly I:C LPS CpG Non-CpG PBS Unchallenged

Page 91: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

77

Figure 6. Relative gene expression of the interferons and the interferon inducible gene

2’-5’-oligoadenylate synthetase (OAS) in the lungs on days 2, 4 and 7 post-infection as

determined by quantitative RT-PCR. Twenty-four hours prior to infection, chickens

received intramuscular injections of poly I:C (400 μg), LPS (100 μg), CpG ODN (50 μg),

non-CpG ODN (50 μg), or PBS. Graphed values represent mean gene expression levels

relative to the house keeping gene β-actin ± standard error. Statistical significance

between treatment groups compared to the infected, PBS treated control group was

calculated using an un-paired students t test, and results were considered statistically

significant if p ≤ 0.05 (*) and p ≤ 0.01 (**), considered statistically significant from the

uninfected PBS control group if p ≤ 0.05 (#), and in the case of CpG ODN, statistically

significant from the non-CpG ODN group if p ≤ 0.05 (~).

IFN- Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

1.010- 8

2.010- 8

3.010- 8

4.010- 8

*

#

*

**

#

#

IFN- Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

1.010- 8

2.010- 8

3.010- 8

4.010- 8

5.010- 8

*

#

*

#**

*

#

#

#

#

*

~

IFN- Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

5.010- 1 0

1.010- 0 9

1.510- 0 9

2.010- 0 9

2.510- 0 9

**#

##

**

*

*

#

*#

# #

~

OAS Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

5.010- 9

1.010- 8

1.510- 8

2.010- 8

2.510- 8

#

*

#

~

DC

BA

Poly I:C LPS CpG Non-CpG PBS Unchallenged

Page 92: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

78

Figure 7. Relative gene expression of the cytokines IL-1β, IL-8 and IL-18 in the lung on

days 2, 4 and 7 post-infection as determined by quantitative RT-PCR. Twenty-four hours

prior to infection, chickens received intramuscular injections of poly I:C (400 μg), LPS

(100 μg), CpG ODN (50 μg), non-CpG ODN (50 μg), or PBS. Graphed values represent

mean gene expression levels relative to the house keeping gene β-actin ± standard error.

Statistical significance between treatment groups compared to the infected, PBS treated

control group was calculated using an un-paired students t test, and results were

considered statistically significant if p ≤ 0.05 (*) and p ≤ 0.01 (**), considered

statistically significant from the uninfected PBS control group if p ≤ 0.05 (#), and in the

case of CpG ODN, statistically significant from the non-CpG ODN group if p ≤ 0.05 (~).

IL-1 Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

2.010 - 1 0

4.010 - 1 0

6.010 - 1 0

8.010 - 1 0

# #

#

*

IL-8 Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

1.010 - 9

2.010 - 9

3.010 - 9

4.010 - 9

5.010 - 9

*

#

IL-18 Lung

Days Post-Infection

Rela

tive E

xp

ressio

n

2 4 70

1.010 - 1 0

2.010 - 1 0

3.010 - 1 0

##

*

#

** *

C

BA

Poly I:C LPS CpG Non-CpG PBS Unchallenged

Page 93: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

79

CHAPTER 4:

General Discussion

Toll like receptor ligands have the potential to directly activate cells of the

immune system thereby enhancing effector functions and the production of cytokines.

Here, we report that administration of ligands for TLRs 4 and 21 could up-regulate a

variety of immune system genes in the spleen. These genes include the pro-inflammatory

cytokines IL-1β and IL-8, as well as those associated with antigen-presentation, namely

MHC-II. Importantly, both ligands induced the up-regulation of cytokines associated with

anti-viral responses, including IFN-γ and, in the case of LPS, but not CpG ODN, IFN-α.

As such, these findings raise the possibility that the TLR-mediated induction of an anti-

viral state may confer immunity to a low pathogenic AIV. Indeed, this was the case, as

the studies in the present thesis found that the prophylactic administration of poly I:C,

LPS and CpG ODN induced a significant reduction in virus shedding, with poly I:C

being the most efficacious of the three. Furthermore, analysis of gene expression in the

spleen and lungs identified IFN-α and IL-8 as correlates of immunity for poly I:C, and

IFN-γ for CpG ODN and LPS.

The administration of TLR ligands prophylactically presents itself as a novel

approach to protect chickens against AIV, potentially addressing the several limitations

of vaccination. For example, such limitations include a limited efficacy in reducing virus

shedding as well as a large time interval (1-2 weeks) between the time of vaccination and

the elicitation of protective immune responses. In the present studies, it was shown that

TLR ligand prophylaxis may address these limitations, as a significant reduction in virus

Page 94: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

80

shedding was observed when infection occurred as soon as 24 hours post-TLR ligand

treatment. This, therefore, raises the possibility that TLR ligand prophylaxis may be

effectively employed in different scenarios. For example, if an outbreak were to occur in

a farm, all chickens in neighbouring farms within a certain radius may be immediately

treated with these ligands to contain the spread. Similarly, if a pen of chickens were to

become infected within a farm, TLR ligands may be administered immediately to all of

the other pens to limit the spread of the virus. For this to occur, it is imperative that TLR

ligands be stockpiled in advance and must be efficacious in conferring immunity to a

broad range of influenza virus subtypes. Indeed, considering the fact TLR ligands may be

stored for at least a year in a conventional freezer, this is certainly feasible. Importantly,

as the immunomodulating effects of these ligands are antigen-independent and non-

specific, it is highly likely that they may be efficacious against several AIV subtypes.

What is promising about TLR ligand prophylaxis is that it does not have to be

mutually exclusive with existing vaccination campaigns. In fact, it is reasonable to

assume that TLR ligand prophylaxis may work synergistically with vaccine induced

immunity to further reduce the amount of virus shedding compared to either treatment

alone. Furthermore, a complementary effect may potentially be observed if TLR ligands

are included in an inactivated whole virus vaccine formulation, as immunity may be

conferred within one day post-vaccination by the TLR ligands, thereby protecting

chickens until seroconversion to the vaccine antigen occurs. In fact, TLR ligands in this

scenario may also act as vaccine adjuvants, thereby inducing even more robust antibody-

mediated and cell-mediated responses (Wang et al., 2009; Mallick et al., 2011). If

vaccination with vaccines containing TLR ligands were to occur in ovo, not only will

Page 95: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

81

administration of these ligands be facilitated through automation, the protective effects

may persist post-hatch as previously demonstrated with respect to other pathogens

(Mackinnon et al., 2009; Taghavi et al., 2009).

For the most part, TLR ligand prophylaxis appears to be safe in many different

species. For example, in humans several different TLR ligands are either approved for

human use or are in phase II/III clinical trials (Kanzler et al., 2007). In chickens, it

appears that this is the case as well, as no detectable signs of distress were demonstrated

in the present studies. Furthermore, several studies have used TLR ligands in vivo in

chickens with no reported signs of toxicity (Dar et al., 2009a; Mallick et al., 2011; Parvizi

et al., 2012). Taken together, it is reasonable to assume that TLR ligand prophylaxis may

be well tolerated in chickens.

Although TLR ligand prophylaxis shows great promise, there are still a few

limitations that need to be addressed. First, it is important to identify ways to enhance the

duration of efficacy of these ligands. It has previously been shown in chickens that the

protective benefits conferred by the ligands is highest when the challenge occurs within 3

days of treatment, and decreases if this time period is extended to 6 days and is absent at

9 days and onwards (Gomis et al., 2003). One potential reason for this limited duration

may be due to the fact that ligands such as CpG ODN and poly I:C may be degraded in

vivo by nucleases. As such, these ligands should be formulated in such a way to render

them resistant to nucleases, for example with phosphorothioate backbones in the case of

CpG ODN (Brown et al., 1994). Similarly, CpG ODN and poly I:C may be encapsulated

in protective vehicles, such as nanoparticles, to prevent extracellular degradation and

facilitate their delivery to endosomes. As an added bonus, these encapsulated ligands

Page 96: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

82

may subsequently be aerosolized (Evans et al., 2011), thereby addressing the second

limitation which is to identify ways to facilitate the administration of these ligands. In the

present studies, ligands were administered through the i.m. route, which is laborious and

time consuming in a commercial setting, and thereby not practical in the face of an AIV

outbreak. However, if these ligands were to be successfully aerosolized while retaining

their immunostimulatory properties, large numbers of birds can be treated simultaneously

within minutes. Lastly, it is also important to consider the cost-benefit aspect of

administering these ligands. For the most part, the costs of these ligands are significantly

high, especially in the case of CpG ODN. Therefore, to encourage widespread adoption

of TLR ligand prophylaxis, a significant reduction in price of these ligands is required.

An underlying question that remains unaddressed in the thesis is whether or not

the protection conferred by these TLR ligands may also extend towards highly

pathogenic avian influenza viruses, namely H5N1. Chickens that have been infected with

low pathogenic strains like H4N6 tend to have few to no signs of illness, and replication

of the virus is typically localized to the cells of the lung and gastrointestinal tract. In

contrast, chickens infected with highly pathogenic strains demonstrate significant signs of

morbidity which most likely leads to mortality, resulting from rapid dissemination of the

virus from the lung and gastrointestinal tract into systemic circulation thereby infecting

numerous organs including the brain and kidneys. As such, the difference in pathogenesis

and sites of replication may serves as obstacles to reduce the efficacy of TLR ligand

treatments against H5N1. Moreover, low pathogenic strains lack many of the virulence

factors and immune evasive mechanisms associated with highly pathogenic strains, such

as the non-structural protein 1, which acts to suppress the host interferon responses.

Page 97: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

83

Furthermore, it has been demonstrated that highly pathogenic AIV are more resistant to

the effects of type I IFNs in chickens (Penski et al., 2011).This presents itself as a

potential problem, as immunity conferred by the TLR ligands in the study presented in

this thesis was associated with an up-regulation of IFN-α and IFN-γ. Importantly, it is

possible that the up-regulation of these cytokines by TLR ligands may in fact exacerbate

the signs of disease and increase the incidence of mortality due to the phenomenon of

‘cytokine storm’ and the induction of deleterious immune responses. Despite that, TLR

ligands have been shown to protect against both low and highly pathogenic strains in

mice, which raises the possibility that TLR ligands may protect chickens against H5N1,

and should therefore be the aims of future research to see if this is the case.

Another factor to consider while interpreting some of the results is the fact that

the changes in cytokine production and other related molecules was demonstrated at the

transcript level and not at the protein level. Although in most cases changes at the

transcript level are indicative of similar changes in the protein level, this is not always

true. For example, some cytokine genes in certain cell subsets may be translationally, and

not transcriptionally regulated, as has been previously shown in murine invariant natural

killer T cells (Nagaleekar et al., 2011). On a similar note, it may also be possible that

cytokines and other anti-microbial molecules may be stored in pre-formed granules to be

released upon TLR stimulation, similar to what has been shown in mammalian platelets

and granulocytes (Semple et al., 2011). As such, the results determined by real-time PCR

may be slightly different than what would be observed if measuring at the protein level.

However, even with these limitations, real-time PCR in chickens is still widely used and

Page 98: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

84

acceptable when analysing cytokine levels, owing in part to the fact that there is a lack of

commercially available antibodies against chicken cytokines and surface markers.

Although we speculate that IL-8 and the IFNs may serve as potential correlates of

immunity for the TLR ligand treatments, it is presently unclear whether this is indeed the

case. It is possible that the changes in gene expression levels observed may not be due to

the TLR ligand treatments, but a result of the large differences in virus load amongst the

treatment groups. However, to properly address this issue would require blocking the

activity of each of these cytokines in vivo, which is possible using the current technology,

such as small interfering RNAs or cytokine neutralizing antibodies, and future studies

should aim at optimizing these techniques in order to fully assess the roles of each

cytokine.

In conclusion, it is clear that TLR ligands may influence the expression of

immune system genes in the spleens of chickens as well as confer immunity to AIV.

Future studies should focus determining the duration of efficacy of these ligands in this

scenario, as well as identifying cost-effective and less labour intensive ways of

administering them. Importantly, future studies should determine whether TLR ligand

prophylaxis may protect against other subtypes of AIV, especially those which are highly

pathogenic.

Page 99: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

85

References

Abasht, B., Kaiser, M.G., Lamont, S.J., 2008. Toll-like receptor gene expression in

cecum and spleen of advanced intercross line chicks infected with Salmonella

enterica serovar Enteritidis. Vet. Immunol. Immunopathol. 123, 314-23.

Abdul-Careem, M.F., Firoz Mian, M., Gillgrass, A.E., Chenoweth, M.J., Barra, N.G.,

Chan, T., Al-Garawi, A.A., Chew, M.V., Yue, G., Roojen, N.V., Xing, Z., Ashkar,

A.A., 2011. FimH, a TLR4 ligand, induces innate antiviral responses in the lung

leading to protection against lethal influenza infection in mice. Antiviral Res.

Abdul-Careem, M.F., Haq, K., Shanmuganathan, S., Read, L.R., Schat, K.A., Heidari,

M., Sharif, S., 2009. Induction of innate host responses in the lungs of chickens

following infection with a very virulent strain of Marek’s disease virus. Virology

393, 250-7.

Abdul-Careem, M.F., Hunter, B.D., Lee, L.F., Fairbrother, J.H., Haghighi, H.R., Read,

L., Parvizi, P., Heidari, M., Sharif, S., 2008a. Host responses in the bursa of

Fabricius of chickens infected with virulent Marek’s disease virus. Virology 379,

256-65.

Abdul-Careem, M.F., Hunter, B.D., Parvizi, P., Haghighi, H.R., Thanthrige-Don, N.,

Sharif, S., 2007. Cytokine gene expression patterns associated with immunization

against Marek’s disease in chickens. Vaccine 25, 424-32.

Abdul-Careem, M.F., Hunter, B.D., Sarson, A.J., Mayameei, A., Zhou, H., Sharif, S.,

2006. Marek’s disease virus-induced transient paralysis is associated with cytokine

gene expression in the nervous system. Viral Immunol. 19, 167-76.

Abdul-Careem, M.F., Hunter, D.B., Lambourne, M.D., Read, L.R., Parvizi, P., Sharif, S.,

2008b. Expression of cytokine genes following pre- and post-hatch immunization of

chickens with herpesvirus of turkeys. Vaccine 26, 2369-77.

Akira, S., Hoshino, K., 2003. Myeloid Differentiation Factor 88–Dependent and –

Independent Pathways in Toll-Like Receptor Signaling. J. Infect. Dis. 187, S356-

S363.

Akira, S., Takeda, K., 2004. Toll-like receptor signalling. Nat. Rev. Immunol. 4, 499-

511.

Akira, S., Takeda, K., Kaisho, T., 2001. Toll-like receptors: critical proteins linking

innate and acquired immunity. Nat. Immunol. 2, 675-80.

Akira, S., Uematsu, S., Takeuchi, O., 2006. Pathogen recognition and innate immunity.

Cell 124, 783-801.

Page 100: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

86

Ameiss, K.A., El Attrache, J., Barri, A., McElroy, A.P., Caldwell, D.J., 2006. Influence

of orally administered CpG-ODNs on the humoral response to bovine serum

albumin (BSA) in chickens. Vet. Immunol. Immunopathol. 110, 257-67.

Ashkar, A.A., Bauer, S., Mitchell, W.J., Vieira, J., Rosenthal, K.L., 2003. Local delivery

of CpG oligodeoxynucleotides induces rapid changes in the genital mucosa and

inhibits replication, but not entry, of herpes simplex virus type 2. J. Virol. 77, 8948-

56.

Balasubramaniam, V.R.M.T., Wai, T.H., Omar, A.R., Othman, I., Hassan, S.S., 2012.

Cellular transcripts of chicken brain tissues in response to H5N1 and Newcastle

disease virus infection. Virol. J. 9, 53.

Barr, T.A., Brown, S., Ryan, G., Zhao, J., Gray, D., 2007. TLR-mediated stimulation of

APC: Distinct cytokine responses of B cells and dendritic cells. Eur. J. Immunol. 37,

3040-53.

Booth, J., Wilson, H., Jimbo, S., Mutwiri, G., 2010. Modulation of B cell responses by

Toll-like receptors. Cell Tissue Res. 131-140.

Brinkmann, V., Geiger, T., Alkan, S., Heusser, C.H., 1993. Interferon alpha increases the

frequency of interferon gamma-producing human CD4+ T cells. J. Exp. Med. 178,

1655-63.

Brisbin, J.T., Gong, J., Parvizi, P., Sharif, S., 2010. Effects of lactobacilli on cytokine

expression by chicken spleen and cecal tonsil cells. Clin. Vaccine Immunol. 17,

1337-43.

Brisbin, J.T., Zhou, H., Gong, J., Sabour, P., Akbari, M.R., Haghighi, H.R., Yu, H.,

Clarke, A., Sarson, A.J., Sharif, S., 2008. Gene expression profiling of chicken

lymphoid cells after treatment with Lactobacillus acidophilus cellular components.

Dev. Comp. Immunol. 32, 563-74.

Brown, C.C., Olander, H.J., Senne, D.A., 1992. A pathogenesis study of highly

pathogenic avian influenza virus H5N2 in chickens, using immunohistochemistry. J.

Comp. Pathol. 107, 341-8.

Brown, D.A., Kang, S.H., Gryaznov, S.M., DeDionisio, L., Heidenreich, O., Sullivan, S.,

Xu, X., Nerenberg, M.I., 1994. Effect of phosphorothioate modification of

oligodeoxynucleotides on specific protein binding. J. Biol. Chem. 269, 26801-5.

Brownlie, R., Zhu, J., Allan, B., Mutwiri, G.K., Babiuk, L.A., Potter, A., Griebel, P.,

2009. Chicken TLR21 acts as a functional homologue to mammalian TLR9 in the

recognition of CpG oligodeoxynucleotides. Mol. Immunol. 46, 3163-70.

Page 101: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

87

Buhtoiarov, I.N., Lum, H., Berke, G., Paulnock, D.M., Sondel, P.M., Rakhmilevich,

A.L., 2005. CD40 ligation activates murine macrophages via an IFN-gamma-

dependent mechanism resulting in tumor cell destruction in vitro. J. Immunol. 174,

6013-22.

Burggraaf, S., Bingham, J., Payne, J., Kimpton, W.G., Lowenthal, J.W., Bean, A.G.D.,

2011. Increased inducible nitric oxide synthase expression in organs is associated

with a higher severity of H5N1 influenza virus infection. PloS One 6, e14561.

Capua, I., Marangon, S., 2006. Control of avian influenza in poultry. Emerg. Infect. Dis.

12, 1319-24.

Chaung, H.-C., Cheng, L.-T., Hung, L.-H., Tsai, P.-C., Skountzou, I., Wang, B.,

Compans, R.W., Lien, Y.-Y., 2011. Salmonella flagellin enhances mucosal

immunity of avian influenza vaccine in chickens. Vet. Microbiol.

Chaussé, A.-M., Grépinet, O., Bottreau, E., Le Vern, Y., Menanteau, P., Trotereau, J.,

Robert, V., Wu, Z., Kerboeuf, D., Beaumont, C., Velge, P., 2011. Expression of toll-

like receptor 4 and downstream effectors in selected cecal cell subpopulations of

chicks resistant or susceptible to salmonella carrier state. Infect. Immun. 79, 3445-

54.

Cheng, Q., Jiang, Z., Xu, C., Li, H., Cao, D., Yang, Z., Cao, G., Linghua, Z., 2010. CpG

oligodeoxynucleotide promotes protective immunity in the enteric mucosa and

suppresses enterotoxigenic E. coli in the weaning piglets. Int. Immunopharmacol.

10, 1249-60.

Chmielewski, R., 2011. Avian Influenza : Public Health and Food Safety Concerns.

Annu. Rev. Food. Sci. Technol.

Chow, J.C., Young, D.W., Golenbock, D.T., Christ, W.J., Gusovsky, F., 1999. Toll-like

receptor-4 mediates lipopolysaccharide-induced signal transduction. J. Biol. Chem.

274, 10689-92.

Ciraci, C., Lamont, S.J., 2011. Avian-specific TLRs and downstream effector responses

to CpG-induction in chicken macrophages. Dev. Comp. Immunol. 35, 392-8.

Cluff, C.W., Baldridge, J.R., Stöver, A.G., Evans, J.T., Johnson, D.A., Lacy, M.J.,

Clawson, V.G., Yorgensen, V.M., Johnson, C.L., Livesay, M.T., Hershberg, R.M.,

Persing, D.H., 2005. Synthetic toll-like receptor 4 agonists stimulate innate

resistance to infectious challenge. Infect. Immun. 73, 3044-52.

Conroy, H., Marshall, N.A., Mills, K.H.G., 2008. TLR ligand suppression or

enhancement of Treg cells? A double-edged sword in immunity to tumours.

Oncogene 27, 168-80.

Page 102: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

88

Dalloul, R.A., Lillehoj, H.S., Okamura, M., Xie, H., Min, W., Ding, X., Heckert, R.A.,

2004. In vivo effects of CpG oligodeoxynucleotide on Eimeria infection in chickens.

Avian Dis. 48, 783-90.

Dar, A., Allan, B., Gomis, S., 2009a. Immunotherapeutic potential of CpG

oligonucleotides in chickens. J. Poult. Sci. 46, 69-80.

Dar, A., Potter, A., Tikoo, S., Gerdts, V., Lai, K., Babiuk, L.A., Mutwiri, G., 2009b. CpG

Oligodeoxynucleotides Activate Innate Immune Response that Suppresses

Infectious Bronchitis Virus Replication in Chicken Embryos. Avian Dis. 53, 261-

267.

De Boever, S., Croubels, S., Meyer, E., Sys, S., Beyaert, R., Ducatelle, R., De Backer, P.,

2009. Characterization of an intravenous lipopolysaccharide inflammation model in

broiler chickens. Avian Pathol. 38, 403-11.

De Geus, E.D., Rebel, J.M.J., Vervelde, L., 2011. Kinetics of the avian influenza-specific

humoral responses in lung are indicative of local antibody production. Dev. Comp.

Immunol.

Degen, W.G.J., Smith, J., Simmelink, B., Glass, E.J., Burt, D.W., Schijns, V.E.J.C., 2006.

Molecular immunophenotyping of lungs and spleens in naive and vaccinated

chickens early after pulmonary avian influenza A (H9N2) virus infection. Vaccine

24, 6096-109.

Doly, J., Civas, A., Navarro, S., Uze, G., 1998. Type I interferons: expression and

signalization. Cell. Mol. Life Sci. 54, 1109-1121.

Dzopalic, T., Dragicevic, A., Vasilijic, S., Vucevic, D., Majstorovic, I., Bozic, B., Balint,

B., Colic, M., 2010. Loxoribine, a selective Toll-like receptor 7 agonist, induces

maturation of human monocyte-derived dendritic cells and stimulates their Th-1-

and Th-17-polarizing capability. Int. Immunopharmacol. 10, 1428-33.

Ehrhardt, C., Seyer, R., Hrincius, E.R., Eierhoff, T., Wolff, T., Ludwig, S., 2010.

Interplay between influenza A virus and the innate immune signaling. Microbes

Infect. 12, 81-7.

Erhard, M.H., Schmidt, P., Zinsmeister, P., Hofmann, A., Münster, U., Kaspers, B.,

Wiesmüller, K.H., Bessler, W.G., Stangassinger, M., 2000. Adjuvant effects of

various lipopeptides and interferon-gamma on the humoral immune response of

chickens. Poult. Sci. 79, 1264-70.

Evans, S.E., Tuvim, M.J., Fox, C.J., Sachdev, N., Gibiansky, L., Dickey, B.F., 2011.

Inhaled innate immune ligands to prevent pneumonia. Br. J. Pharmacol. 163, 195-

206.

Page 103: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

89

Ewald, S.J., Kapczynski, D.R., Livant, E.J., Suarez, D.L., Ralph, J., McLeod, S., Miller,

C., 2011. Association of Mx1 Asn631 variant alleles with reductions in morbidity,

early mortality, viral shedding, and cytokine responses in chickens infected with a

highly pathogenic avian influenza virus. Immunogenetics 63, 363-75.

Ferdous, F., Maurice, D., Scott, T., 2008. Broiler chick thrombocyte response to

lipopolysaccharide. Poult. Sci. 87, 61-3.

Flores, R.R., Diggs, K.A., Tait, L.M., Morel, P.A., 2007. IFN-gamma negatively

regulates CpG-induced IL-10 in bone marrow-derived dendritic cells. J. Immunol.

178, 211-8.

Fukui, A., Inoue, N., Matsumoto, M., Nomura, M., Yamada, K., Matsuda, Y.,

Toyoshima, K., Seya, T., 2001. Molecular cloning and functional characterization of

chicken toll-like receptors. A single chicken toll covers multiple molecular patterns.

J. Biol. Chem. 276, 47143-9.

Gaudreault, E., Gosselin, J., 2008. Leukotriene B4 induces release of antimicrobial

peptides in lungs of virally infected mice. J. Immunol. 180, 6211-21.

Genovese, K.J., He, H., Lowry, V.K., Nisbet, D.J., Kogut, M.H., 2007. Dynamics of the

avian inflammatory response to Salmonella following administration of the Toll-like

receptor 5 agonist flagellin. FEMS Immunol. Med. Microbiol. 51, 112-7.

Ghosh, T.K., Mickelson, D.J., Solberg, J.C., Lipson, K.E., Inglefield, J.R., Alkan, S.S.,

2007. TLR-TLR cross talk in human PBMC resulting in synergistic and antagonistic

regulation of type-1 and 2 interferons, IL-12 and TNF-alpha. Int.

Immunopharmacol. 7, 1111-21.

Gomis, S., Babiuk, L., Allan, B., Willson, P., Waters, E., Ambrose, N., Hecker, R.,

Potter, A., 2004. Protection of neonatal chicks against a lethal challenge of

Escherichia coli using DNA containing cytosine-phosphodiester-guanine motifs.

Avian Dis. 48, 813-22.

Gomis, S., Babiuk, L., Godson, D.L., Allan, B., Thrush, T., Townsend, H., Willson, P.,

Waters, E., Hecker, R., Potter, A., 2003. Protection of chickens against Escherichia

coli infections by DNA containing CpG motifs. Infect. Immun. 71, 857-63.

Guillot, L., Le Goffic, R., Bloch, S., Escriou, N., Akira, S., Chignard, M., Si-Tahar, M.,

2005. Involvement of toll-like receptor 3 in the immune response of lung epithelial

cells to double-stranded RNA and influenza A virus. J. Biol. Chem. 280, 5571-80.

Guo, H., Kumar, P., Malarkannan, S., 2011. Evasion of natural killer cells by influenza

virus. J. Leukocyte Biol. 89, 189-94.

Page 104: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

90

Haghighi, H.R., Read, L.R., Haeryfar, S.M.M., Behboudi, S., Sharif, S., 2009.

Identification of a dual-specific T cell epitope of the hemagglutinin antigen of an h5

avian influenza virus in chickens. PloS One 4, e7772.

Haghihghi, H.R., Read, L.R., Mohammadi, H., Pei, Y., Ursprung, C., Nagy, E.,

Behboudi, S., Haeryfar, S.M.M., Sharif, S., 2010. Characterization of host responses

against a recombinant fowlpox virus-vectored vaccine expressing the hemagglutinin

antigen of an avian influenza virus. Clin. Vaccine Immunol. 17, 454-63.

Hanten, J.A., Vasilakos, J.P., Riter, C.L., Neys, L., Lipson, K.E., Alkan, S.S., Birmachu,

W., 2008. Comparison of human B cell activation by TLR7 and TLR9 agonists.

BMC Immunol. 9, 39.

Haq, K., Abdul-Careem, M.F., Shanmuganthan, S., Thanthrige-Don, N., Read, L.R.,

Sharif, S., 2010. Vaccine-induced host responses against very virulent Marek’s

disease virus infection in the lungs of chickens. Vaccine 28, 5565-72.

Hartmann, G., Krieg, A.M., 1999. CpG DNA and LPS induce distinct patterns of

activation in human monocytes. Gene Ther. 6, 893-903.

He, H., Crippen, T.L., Farnell, M.B., Kogut, M.H., 2003. Identification of CpG

oligodeoxynucleotide motifs that stimulate nitric oxide and cytokine production in

avian macrophage and peripheral blood mononuclear cells. Dev. Comp. Immunol.

27, 621-627.

He, H., Genovese, K.J., Kogut, M.H., 2011a. Modulation of chicken macrophage effector

function by T(H)1/T(H)2 cytokines. Cytokine 1-7.

He, H., Genovese, K.J., Nisbet, D.J., Kogut, M.H., 2006. Profile of Toll-like receptor

expressions and induction of nitric oxide synthesis by Toll-like receptor agonists in

chicken monocytes. Mol. Immunol. 43, 783-9.

He, H., Genovese, K.J., Nisbet, D.J., Kogut, M.H., 2007a. Synergy of CpG

oligodeoxynucleotide and double-stranded RNA (poly I:C) on nitric oxide induction

in chicken peripheral blood monocytes. Mol. Immunol. 44, 3234-42.

He, H., Genovese, K.J., Swaggerty, C.L., Mackinnon, K.M., Kogut, M.H., 2012. Co-

stimulation with TLR3 and TLR21 ligands synergistically up-regulates Th1-

cytokine IFN-γ and regulatory cytokine IL-10 expression in chicken monocytes.

Dev. Comp. Immunol. 36, 756-60.

He, H., Genovese, K.J., Swaggerty, C.L., Nisbet, D.J., Kogut, M.H., 2007b. In vivo

priming heterophil innate immune functions and increasing resistance to Salmonella

enteritidis infection in neonatal chickens by immune stimulatory CpG

oligodeoxynucleotides. Vet. Immunol. Immunopathol. 117, 275-83.

Page 105: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

91

He, H., MacKinnon, K.M., Genovese, K.J., Kogut, M.H., 2011b. CpG

oligodeoxynucleotide and double-stranded RNA synergize to enhance nitric oxide

production and mRNA expression of inducible nitric oxide synthase, pro-

inflammatory cytokines and chemokines in chicken monocytes. Innate Immun. 17,

137-44.

Hopkins, P.A., Sriskandan, S., 2005. Mammalian Toll-like receptors: to immunity and

beyond. Clin. Exp. Immunol. 140, 395-407.

Ichinohe, T., Watanabe, I., Ito, S., Fujii, H., Moriyama, M., Tamura, S.-ichi, Takahashi,

H., Sawa, H., Chiba, J., Kurata, T., Sata, T., Hasegawa, H., 2005. Synthetic double-

stranded RNA poly(I:C) combined with mucosal vaccine protects against influenza

virus infection. J. Virol. 79, 2910-9.

Iqbal, M., Philbin, V.J., Smith, A.L., 2005. Expression patterns of chicken Toll-like

receptor mRNA in tissues, immune cell subsets and cell lines. Vet. Immunol.

Immunopathol. 104, 117-27.

Jenkins, K.A., Lowenthal, J.W., Kimpton, W., Bean, A.G.D., 2009. The in vitro and in

ovo responses of chickens to TLR9 subfamily ligands. Dev. Comp. Immunol. 33,

660-7.

Jiang, H., Yang, H., Kapczynski, D.R., 2011. Chicken interferon alpha pretreatment

reduces virus replication of pandemic H1N1 and H5N9 avian influenza viruses in

lung cell cultures from different avian species. Virol. J. 8, 447.

Jiang, W., Lederman, M.M., Harding, C.V., Rodriguez, B., Mohner, R.J., Sieg, S.F.,

2007a. TLR9 stimulation drives naïve B cells to proliferate and to attain enhanced

antigen presenting function. Eur. J. Immunol. 37, 2205-13.

Jiang, Y., Yu, K., Zhang, H., Zhang, P., Li, C., Tian, G., Li, Y., Wang, X., Ge, J., Bu, Z.,

Chen, H., 2007b. Enhanced protective efficacy of H5 subtype avian influenza DNA

vaccine with codon optimized HA gene in a pCAGGS plasmid vector. Antiviral Res.

75, 234-41.

Kanzler, H., Barrat, F.J., Hessel, E.M., Coffman, R.L., 2007. Therapeutic targeting of

innate immunity with Toll-like receptor agonists and antagonists. Nat. Med. 13, 552-

9.

Karpala, A.J., Bingham, J., Schat, K.A., Chen, L.-M., Donis, R.O., Lowenthal, J.W.,

Bean, A.G.D., 2011a. Highly pathogenic (H5N1) avian influenza induces an

inflammatory T helper type 1 cytokine response in the chicken. J. Interferon

Cytokine Res. 31, 393-400.

Karpala, A.J., Lowenthal, J.W., Bean, A.G., 2008. Activation of the TLR3 pathway

regulates IFNbeta production in chickens. Dev. Comp. Immunol. 32, 435-44.

Page 106: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

92

Karpala, A.J., Stewart, C., McKay, J., Lowenthal, J.W., Bean, A.G.D., 2011b.

Characterization of chicken Mda5 activity: regulation of IFN-β in the absence of

RIG-I functionality. J. Immunol. 186, 5397-405.

Kawai, T., Akira, S., 2010. The role of pattern-recognition receptors in innate immunity:

update on Toll-like receptors. Nat. Immunol. 11, 373-84.

Keestra, A.M., de Zoete, M.R., Bouwman, L.I., van Putten, J.P.M., 2010. Chicken

TLR21 is an innate CpG DNA receptor distinct from mammalian TLR9. J.

Immunol. 185, 460-7.

Keestra, A.M., van Putten, J.P.M., 2008. Unique properties of the chicken TLR4/MD-2

complex: selective lipopolysaccharide activation of the MyD88-dependent pathway.

J. Immunol. 181, 4354-62.

Kindt, T.J., Goldsby, R.A., Osborne, B.A., Kuby, J., 2006. Kuby Immunology 6th

Edition. W.H. Freeman & Company.

Kochs, G., García-Sastre, A., Martínez-Sobrido, L., 2007. Multiple anti-interferon actions

of the influenza A virus NS1 protein. J. Virol. 81, 7011-21.

Kodihalli, S., Haynes, J.R., Robinson, H.L., Webster, R.G., 1997. Cross-protection

among lethal H5N2 influenza viruses induced by DNA vaccine to the

hemagglutinin. J. Virol. 71, 3391-6.

Kogut, M.H., 2002. Dynamics of a protective avian inflammatory response: the role of an

IL-8-like cytokine in the recruitment of heterophils to the site of organ invasion by

Salmonella enteritidis. Comp. Immunol. Microbiol. Infect. Dis. 25, 159-72.

Kogut, M.H., He, H., Kaiser, P., 2005a. Lipopolysaccharide binding protein/CD14/

TLR4-dependent recognition of salmonella LPS induces the functional activation of

chicken heterophils and up-regulation of pro-inflammatory cytokine and chemokine

gene expression in these cells. Anim. Biotechnol. 16, 165-81.

Kogut, M.H., Iqbal, M., He, H., Philbin, V., Kaiser, P., Smith, A., 2005b. Expression and

function of Toll-like receptors in chicken heterophils. Dev. Comp. Immunol. 29,

791-807.

Krug, A., Rothenfusser, S., Hornung, V., Jahrsdörfer, B., Blackwell, S., Ballas, Z.K.,

Endres, S., Krieg, A.M., Hartmann, G., 2001. Identification of CpG oligonucleotide

sequences with high induction of IFN-alpha/beta in plasmacytoid dendritic cells.

Eur. J. Immunol. 31, 2154-63.

Kugel, D., Kochs, G., Obojes, K., Roth, J., Kobinger, G.P., Kobasa, D., Haller, O.,

Staeheli, P., von Messling, V., 2009. Intranasal administration of alpha interferon

reduces seasonal influenza A virus morbidity in ferrets. J. Virol. 83, 3843-51.

Page 107: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

93

Kulkarni, R., Behboudi, S., Sharif, S., 2010. Insights into the role of Toll-like receptors in

modulation of T cell responses. Cell Tissue Res. 141-152.

Le Gall-Reculé, G., Cherbonnel, M., Pelotte, N., Blanchard, P., Morin, Y., Jestin, V.,

2007. Importance of a prime-boost DNA/protein vaccination to protect chickens

against low-pathogenic H7 avian influenza infection. Avian Dis. 51, 490-4.

Le Goffic, R., Pothlichet, J., Vitour, D., Fujita, T., Meurs, E., Chignard, M., Si-Tahar, M.,

2007. Cutting Edge: Influenza A virus activates TLR3-dependent inflammatory and

RIG-I-dependent antiviral responses in human lung epithelial cells. J. Immunol. 178,

3368-72.

Lee, C.-W., Suarez, D.L., 2005. Avian influenza virus: prospects for prevention and

control by vaccination. Anim. Health Res. Rev. 6, 1-15.

Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J.M., Hoffmann, J. a, 1996. The

dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent

antifungal response in Drosophila adults. Cell 86, 973-83.

Lenert, P., Brummel, R., Field, E.H., Ashman, R.F., 2005. TLR-9 activation of marginal

zone B cells in lupus mice regulates immunity through increased IL-10 production.

J. Clin. Immunol. 25, 29-40.

Leveque, G., Forgetta, V., Morroll, S., Smith, A.L., Bumstead, N., Barrow, P., Morgan,

K., Malo, D., Loredo-Osti, J.C., 2003. Allelic Variation in TLR4 Is Linked to

Susceptibility to Salmonella enterica Serovar Typhimurium Infection in Chickens.

Infect. Immun. 71, 1116-1124.

Lian, L., Ciraci, C., Chang, G., Hu, J., Lamont, S.J., 2012. NLRC5 knockdown in

chicken macrophages alters response to LPS and poly (I:C) stimulation. BMC Vet.

Res. 8, 23.

Liang, S.-L., Quirk, D., Zhou, A., 2006. RNase L: its biological roles and regulation.

IUBMB Life 58, 508-14.

Lin, Y.P., Shaw, M., Gregory, V., Cameron, K., Lim, W., Klimov, A., Subbarao, K.,

Guan, Y., Krauss, S., Shortridge, K., Webster, R., Cox, N., Hay, A., 2000. Avian-to-

human transmission of H9N2 subtype influenza A viruses: relationship between

H9N2 and H5N1 human isolates. Proc. Natl. Acad. Sci. U. S. A. 97, 9654-8.

Linghua, Z., Xingshan, T., Fengzhen, Z., 2007. Vaccination with Newcastle disease

vaccine and CpG oligodeoxynucleotides induces specific immunity and protection

against Newcastle disease virus in SPF chicken. Vet. Immunol. Immunopathol. 115,

216-22.

Page 108: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

94

Liniger, M., Summerfield, A., Zimmer, G., McCullough, K.C., Ruggli, N., 2012. Chicken

cells sense influenza A virus infection through MDA5 and CARDIF signaling

involving LGP2. J. Virol. 86, 705-17.

Liu, Y.C., Gray, R.C., Hardy, G.A.D., Kuchtey, J., Abbott, D.W., Emancipator, S.N.,

Harding, C.V., 2010. CpG-B oligodeoxynucleotides inhibit TLR-dependent and -

independent induction of type I IFN in dendritic cells. J. Immunol. 184, 3367-76.

Lu, Y., Sarson, A.J., Gong, J., Zhou, H., Zhu, W., Kang, Z., Yu, H., Sharif, S., Han, Y.,

2009. Expression profiles of genes in Toll-like receptor-mediated signaling of

broilers infected with Clostridium perfringens. Clin. Vaccine Immunol. 16, 1639-47.

Mackinnon, K.M., He, H., Swaggerty, C.L., McReynolds, J.L., Genovese, K.J., Duke,

S.E., Nerren, J.R., Kogut, M.H., 2009. In ovo treatment with CpG

oligodeoxynucleotides decreases colonization of Salmonella enteriditis in broiler

chickens. Vet. Immunol. Immunopathol. 127, 371-5.

Mallick, A.I., Kulkarni, R.R., St Paul, M., Parvizi, P., Nagy, E., Behboudi, S., Sharif, S.,

2012. Vaccination with CpG-Adjuvanted Avian Influenza Virosomes Promotes

Antiviral Immune Responses and Reduces Virus Shedding in Chickens. Viral

Immunol. 25, 226-31.

Mallick, A.I., Parvizi, P., Read, L.R., Nagy, E., Behboudi, S., Sharif, S., 2011.

Enhancement of immunogenicity of a virosome-based avian influenza vaccine in

chickens by incorporating CpG-ODN. Vaccine 29, 1657-65.

Marcus, P.I., Sekellick, M.J., 2001. Combined sequential treatment with interferon and

dsRNA abrogates virus resistance to interferon action. J. Interferon Cytokine Res.

21, 423-9.

Marie, C., Pitton, C., Fitting, C., Cavaillon, J.M., 1996. Regulation by anti-inflammatory

cytokines (IL-4, IL-10, IL-13, TGFbeta)of interleukin-8 production by LPS- and/ or

TNFalpha-activated human polymorphonuclear cells. Mediators Inflamm. 5, 334-40.

Matsumoto, M., Seya, T., 2008. TLR3: interferon induction by double-stranded RNA

including poly(I:C). Adv. Drug Deliv. Rev. 60, 805-12.

Medzhitov, R., 2001. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1,

135-45.

Medzhitov, R., Janeway, C.A., 1997. Innate immunity: the virtues of a nonclonal system

of recognition. Cell 91, 295-8.

Medzhitov, R., Preston-Hurlburt, P., Janeway, C. a, 1997. A human homologue of the

Drosophila Toll protein signals activation of adaptive immunity. Nature 388, 394-7.

Page 109: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

95

Meng, S., Yang, L., Xu, C., Qin, Z., Xu, H., Wang, Y., Sun, L., Liu, W., 2011.

Recombinant chicken interferon-α inhibits H9N2 avian influenza virus replication in

vivo by oral administration. J. Interferon Cytokine Res. 31, 533-8.

Min, J.-Y., Krug, R.M., 2006. The primary function of RNA binding by the influenza A

virus NS1 protein in infected cells: Inhibiting the 2’-5' oligo (A) synthetase/RNase L

pathway. Proc. Natl. Acad. Sci. U. S. A. 103, 7100-5.

Mosser, D.M., 2003. The many faces of macrophage activation. J. Leukocyte Biol. 73,

209-212.

Moynagh, P.N., 2005. TLR signalling and activation of IRFs: revisiting old friends from

the NF-kappaB pathway. Trends Immunol. 26, 469-76.

Nagaleekar, V.K., Sabio, G., Aktan, I., Chant, A., Howe, I.W., Thornton, T.M., Benoit,

P.J., Davis, R.J., Rincon, M., Boyson, J.E., 2011. Translational control of NKT cell

cytokine production by p38 MAPK. J. Immunol. 186, 4140-6.

Nang, N.T., Lee, J.S., Song, B.M., Kang, Y.M., Kim, H.S., Seo, S.H., 2011. Induction of

inflammatory cytokines and toll-like receptors in chickens infected with avian H9N2

influenza virus. Vet. Res. 42, 64.

Nguyen-Van-Tam, J.S., Nair, P., Acheson, P., Baker, A., Barker, M., Bracebridge, S.,

Croft, J., Ellis, J., Gelletlie, R., Gent, N., Ibbotson, S., Joseph, C., Mahgoub, H.,

Monk, P., Reghitt, T.W., Sundkvist, T., Sellwood, C., Simpson, J., Smith, J.,

Watson, J.M., Zambon, M., Lightfoot, N., 2006. Outbreak of low pathogenicity

H7N3 avian influenza in UK, including associated case of human conjunctivitis.

Euro Surveill. 11, 2952.

Nichani, A.K., Dar, M.A., Mirakhur, K.K., Krieg, A.M., Booth, J.S., Townsend, H.G.G.,

Potter, A.A., Babiuk, L.A., Mutwiri, G.K., 2010. Subcutaneous, but not intratracheal

administration of the TLR9 agonist, CpG DNA transiently reduces parainfluenza-3

virus shedding in newborn lambs. Comp. Immunol. Microbiol. Infect. Dis. 33, e111-

7.

Nichani, A.K., Mena, A., Kaushik, R.S., Mutwiri, G.K., Townsend, H.G.G., Hecker, R.,

Krieg, A.M., Babiuk, L.A., Griebel, P.J., 2006. Stimulation of innate immune

responses by CpG oligodeoxynucleotide in newborn lambs can reduce bovine

herpesvirus-1 shedding. Oligonucleotides 16, 58-67.

Nogusa, S., Ritz, B.W., Kassim, S.H., Jennings, S.R., Gardner, E.M., 2008.

Characterization of age-related changes in natural killer cells during primary

influenza infection in mice. Mech. Ageing Dev. 129, 223-30.

Page 110: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

96

Okamatsu, M., Saito, T., Yamamoto, Y., Mase, M., Tsuduku, S., Nakamura, K.,

Tsukamoto, K., Yamaguchi, S., 2007. Low pathogenicity H5N2 avian influenza

outbreak in Japan during the 2005-2006. Vet. Microbiol. 124, 35-46.

Pantin-Jackwood, M.J., Swayne, D.E., 2009. Pathogenesis and pathobiology of avian

influenza virus infection in birds. Rev. Sci. Tech. 28, 113-36.

Parvizi, P., Mallick, A.I., Haq, K., Schlegel, B., Sharif, S., 2012. A Toll-like receptor 3

agonist (polyI:C) elicits innate host responses in the spleen and lungs of chickens.

Can. J. Vet. Res.

Patel, B.A., Gomis, S., Dar, A., Willson, P.J., Babiuk, L.A., Potter, A., Mutwiri, G.,

Tikoo, S.K., 2008. Oligodeoxynucleotides containing CpG motifs (CpG-ODN)

predominantly induce Th1-type immune response in neonatal chicks. Dev. Comp.

Immunol. 32, 1041-9.

Penski, N., Härtle, S., Rubbenstroth, D., Krohmann, C., Ruggli, N., Schusser, B., Pfann,

M., Reuter, A., Gohrbandt, S., Hundt, J., Veits, J., Breithaupt, A., Kochs, G., Stech,

J., Summerfield, A., Vahlenkamp, T., Kaspers, B., Staeheli, P., 2011. Highly

pathogenic avian influenza viruses do not inhibit interferon synthesis in infected

chickens but can override the interferon-induced antiviral state. J. Virol. 85, 7730-

41.

Pfaffl, M.W., 2001. A new mathematical model for relative quantification in real-time

RT-PCR. Nucleic Acids Res. 29, e45.

Pfaffl, M.W., Horgan, G.W., Dempfle, L., 2002. Relative expression software tool

(REST) for group-wise comparison and statistical analysis of relative expression

results in real-time PCR. Nucleic Acids Res. 30, e36.

Pulendran, B., Kumar, P., Cutler, C.W., Mohamadzadeh, M., Van Dyke, T., Banchereau,

J., 2001. Lipopolysaccharides from distinct pathogens induce different classes of

immune responses in vivo. J. Immunol. 167, 5067-76.

Rao, S., Kong, W.-P., Wei, C.-J., Yang, Z.-Y., Nason, M., Styles, D., DeTolla, L.J.,

Panda, A., Sorrell, E.M., Song, H., Wan, H., Ramirez-Nieto, G.C., Perez, D., Nabel,

G.J., 2008. Multivalent HA DNA vaccination protects against highly pathogenic

H5N1 avian influenza infection in chickens and mice. PloS One 3, e2432.

Rappuoli, R., Del Giudice, G., 2008. Influenza Vaccines For The Future. Birkhauser

Verlag.

Rebel, J.M., Peeters, B., Fijten, H., Post, J., Cornelissen, J., Vervelde, L., 2011. Highly

pathogenic or low pathogenic avian influenza virus subtype H7N1 infection in

chicken lungs: small differences in general acute responses. Vet. Res. 42, 10.

Page 111: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

97

Renis, H.E., 1970. Effect of poly I: C on experimental respiratory infection in hamsters.

Appl. Microbiol. 20, 821-4.

Report, S., 2005. The 1918 flu virus is resurrected. Nature 437, 794-5.

Reynolds, D., 2006. Update on low pathogenic avian influenza. Vet. Rec. 158, 706-706.

Roach, J.C., Glusman, G., Rowen, L., Kaur, A., Purcell, M.K., Smith, K.D., Hood, L.E.,

Aderem, A., 2005. The evolution of vertebrate Toll-like receptors. Proc. Natl. Acad.

Sci. U. S. A. 102, 9577-82.

Samji, T., 2009. Influenza A: understanding the viral life cycle. Yale J. Biol. Med. 82,

153-9.

Samuel, C.E., 2001. Antiviral actions of interferons. Clin. Microbiol. Rev. 14, 778-809.

Sang, Y., Ross, C.R., Rowland, R.R.R., Blecha, F., 2008. Toll-like receptor 3 activation

decreases porcine arterivirus infection. Viral Immunol. 21, 303-13.

Saraiva, M., O’Garra, A., 2010. The regulation of IL-10 production by immune cells.

Nat. Rev. Immunol. 10, 170-81.

Sarson, A.J., Read, L.R., Haghighi, H.R., Lambourne, M.D., Brisbin, J.T., Zhou, H.,

Sharif, S., 2007. Construction of a microarray specific to the chicken immune

system: profiling gene expression in B cells after lipopolysaccharide stimulation.

Can. J. Vet. Res. 71, 108-18.

Schwarz, H., Schneider, K., Ohnemus, A., Lavric, M., Kothlow, S., Bauer, S., Kaspers,

B., Staeheli, P., 2007. Chicken toll-like receptor 3 recognizes its cognate ligand

when ectopically expressed in human cells. J. Interferon Cytokine Res. 27, 97-101.

Scott, T., Owens, M.D., 2008. Thrombocytes respond to lipopolysaccharide through Toll-

like receptor-4, and MAP kinase and NF-kappaB pathways leading to expression of

interleukin-6 and cyclooxygenase-2 with production of prostaglandin E2. Mol.

Immunol. 45, 1001-8.

Semple, J.W., Italiano, J.E., Freedman, J., 2011. Platelets and the immune continuum.

Nat. Rev. Immunol. 11, 264-74.

Senn, J.J., Burel, S., Henry, S.P., 2005. Non-CpG-containing antisense 2’-methoxyethyl

oligonucleotides activate a proinflammatory response independent of Toll-like

receptor 9 or myeloid differentiation factor 88. J. Pharmacol. Exp. Ther. 314, 972-9.

Seo, S.H., Peiris, M., Webster, R.G., 2002. Protective cross-reactive cellular immunity to

lethal A/Goose/Guangdong/1/96-like H5N1 influenza virus is correlated with the

Page 112: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

98

proportion of pulmonary CD8(+) T cells expressing gamma interferon. J. Virol. 76,

4886-90.

Seo, S.H., Webster, R.G., 2001. Cross-reactive, cell-mediated immunity and protection of

chickens from lethal H5N1 influenza virus infection in Hong Kong poultry markets.

J. Virol. 75, 2516-25.

Shinya, K., Ito, M., Makino, A., Tanaka, M., Miyake, K., Eisfeld, A.J., Kawaoka, Y.,

2012. The TLR4-TRIF pathway protects against H5N1 influenza virus infection. J.

Virol. 86, 19-24.

Shinya, K., Okamura, T., Sueta, S., Kasai, N., Tanaka, M., Ginting, T.E., Makino, A.,

Eisfeld, A.J., Kawaoka, Y., 2011. Toll-like receptor pre-stimulation protects mice

against lethal infection with highly pathogenic influenza viruses. Virol. J. 8, 97.

Sijben, J.W.., Klasing, K.C., Schrama, J.W., Parmentier, H.K., van der Poel, J.J.,

Savelkoul, H.F.., Kaiser, P., 2003. Early in vivo cytokine genes expression in

chickens after challenge with Salmonella typhimurium lipopolysaccharide and

modulation by dietary n−3 polyunsaturated fatty acids. Dev. Comp. Immunol. 27,

611-619.

Slemons, R.D., Swayne, D.E., 2010. Tissue tropism and replicative properties of

waterfowl-origin influenza viruses in chickens. Avian Dis. 39, 521-7.

Spackman, E., Gelb, J., Preskenis, L. a, Ladman, B.S., Pope, C.R., Pantin-Jackwood,

M.J., McKinley, E.T., 2010. The pathogenesis of low pathogenicity H7 avian

influenza viruses in chickens, ducks and turkeys. Virol. J. 7, 331.

St Paul, M., Mallick, A.I., Haq, K., Orouji, S., Abdul-Careem, M.F., Sharif, S., 2011. In

vivo administration of ligands for chicken toll-like receptors 4 and 21 induces the

expression of immune system genes in the spleen. Vet. Immunol. Immunopathol.

144, 228-37.

Stewart, C.R., Bagnaud-Baule, A., Karpala, A.J., Lowther, S., Mohr, P.G., Wise, T.G.,

Lowenthal, J.W., Bean, A.G., 2011a. Toll-Like Receptor 7 Ligands Inhibit Influenza

A Infection in Chickens. J. Interferon Cytokine Res. 00.

Stewart, C.R., Karpala, A.J., Lowther, S., Lowenthal, J.W., Bean, A.G., 2011b.

Immunostimulatory motifs enhance antiviral siRNAs targeting highly pathogenic

avian influenza H5N1. PloS One 6, e21552.

Suarez, D.L., 2010. Avian influenza: our current understanding. Anim. Health Res. Rev.

11, 19-33.

Suarez, D.L., Schultz-Cherry, S., 2000. Immunology of avian influenza virus: a review.

Dev. Comp. Immunol. 24, 269-83.

Page 113: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

99

Szretter, K.J., Balish, A.L., Katz, J.M., 2006. Influenza: propagation, quantification, and

storage. Curr. Protoc. Microbiol. Chapter 15, Unit 15G.1.

Taghavi, A., Allan, B., Mutwiri, G., Foldvari, M., Van Kessel, A., Willson, P., Babiuk,

L., Potter, A., Gomis, S., 2009. Enhancement of immunoprotective effect of CpG-

ODN by formulation with polyphosphazenes against E. coli septicemia in neonatal

chickens. Curr. Drug Delivery 6, 76-82.

Taghavi, A., Allan, B., Mutwiri, G., Van Kessel, A., Willson, P., Babiuk, L., Potter, A.,

Gomis, S., 2008. Protection of neonatal broiler chicks against Salmonella

Typhimurium septicemia by DNA containing CpG motifs. Avian Dis. 52, 398-406.

Tanaka, T., Sunden, Y., Sakoda, Y., Kida, H., Ochiai, K., Umemura, T., 2010.

Lipopolysaccharide treatment and inoculation of influenza A virus results in

influenza virus-associated encephalopathy-like changes in neonatal mice. J.

Neurovirol. 16, 125-32.

Tate, M.D., Brooks, A.G., Reading, P.C., 2008. The role of neutrophils in the upper and

lower respiratory tract during influenza virus infection of mice. Respir. Res. 9, 57.

Tate, M.D., Brooks, A.G., Reading, P.C., Mintern, J.D., 2011. Neutrophils sustain

effective CD8(+) T-cell responses in the respiratory tract following influenza

infection. Immunol. Cell Biol. 1-9.

Temperley, N.D., Berlin, S., Paton, I.R., Griffin, D.K., Burt, D.W., 2008. Evolution of

the chicken Toll-like receptor gene family: a story of gene gain and gene loss. BMC

Genomics 9, 62.

Thanthrige-Don, N., Read, L.R., Abdul-Careem, M.F., Mohammadi, H., Mallick, A.I.,

Sharif, S., 2010. Marek’s disease virus influences the expression of genes associated

with IFN-gamma-inducible MHC class II expression. Viral Immunol. 23, 227-32.

Tseng, L.-P., Chiou, C.-J., Chen, C.-C., Deng, M.-C., Chung, T.-W., Huang, Y.-Y., Liu,

D.-Z., 2009. Effect of lipopolysaccharide on intranasal administration of liposomal

Newcastle disease virus vaccine to SPF chickens. Vet. Immunol. Immunopathol.

131, 285-9.

Uchida, Y., Watanabe, C., Takemae, N., Hayashi, T., Oka, T., Ito, T., Saito, T., 2012.

Identification of host genes linked with the survivability of chickens infected with

recombinant viruses possessing H5N1 surface antigens from a highly pathogenic

avian influenza virus. J. Virol. 86, 2686-95.

Ucla, C., Roux-Lombard, P., Fey, S., Dayer, J.M., Mach, B., 1990. Interferon gamma

drastically modifies the regulation of interleukin 1 genes by endotoxin in U937 cells.

J. Clin. Invest. 85, 185-91.

Page 114: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

100

Ulmer, J.B., Fu, T.M., Deck, R.R., Friedman, A., Guan, L., DeWitt, C., Liu, X., Wang,

S., Liu, M.A., Donnelly, J.J., Caulfield, M.J., 1998. Protective CD4+ and CD8+ T

cells against influenza virus induced by vaccination with nucleoprotein DNA. J.

Virol. 72, 5648-53.

Villanueva, A.I., Kulkarni, R.R., Sharif, S., 2011. Synthetic double-stranded RNA

oligonucleotides are immunostimulatory for chicken spleen cells. Dev. Comp.

Immunol. 35, 28-34.

Vleugels, B., Ververken, C., Goddeeris, B.M., 2002. Stimulatory effect of CpG

sequences on humoral response in chickens. Poult. Sci. 81, 1317-21.

Vollmer, J., Weeratna, R., Payette, P., Jurk, M., Schetter, C., Laucht, M., Wader, T.,

Tluk, S., Liu, M., Davis, H.L., Krieg, A.M., 2004. Characterization of three CpG

oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur. J.

Immunol. 34, 251-62.

WHO, 2002. World Health Organization Manual on Animal Influenza Diagnosis and

Surveillance [WWW Document]. URL

http://www.who.int/csr/resources/publications/influenza/whocdscsrncs20025rev.pdf

Waag, D.M., McCluskie, M.J., Zhang, N., Krieg, A.M., 2006. A CpG oligonucleotide can

protect mice from a low aerosol challenge dose of Burkholderia mallei. Infect.

Immun. 74, 1944-8.

Wang, B.-Z., Xu, R., Quan, F.-S., Kang, S.-M., Wang, L., Compans, R.W., 2010.

Intranasal immunization with influenza VLPs incorporating membrane-anchored

flagellin induces strong heterosubtypic protection. PloS One 5, e13972.

Wang, J.P., Bowen, G.N., Padden, C., Cerny, A., Finberg, R.W., Newburger, P.E., Kurt-

Jones, E.A., 2008. Toll-like receptor-mediated activation of neutrophils by influenza

A virus. Blood 112, 2028-34.

Wang, Y., Shan, C., Ming, S., Liu, Y., Du, Y., Jiang, G., 2009. Immunoadjuvant effects

of bacterial genomic DNA and CpG oligodeoxynucleotides on avian influenza virus

subtype H5N1 inactivated oil emulsion vaccine in chicken. Res. Vet. Sci. 86, 399-

405.

Watson, M.L., White, A.M., Campbell, E.M., Smith, A.W., Uddin, J., Yoshimura, T.,

Westwick, J., 1999. Anti-inflammatory actions of interleukin-13: suppression of

tumor necrosis factor-alpha and antigen-induced leukocyte accumulation in the

guinea pig lung. Am. J. Respir. Cell Mol. Biol. 20, 1007-12.

Wattrang, E., 2009. Phosphorothioate oligodeoxyribonucleotides induce in vitro

proliferation of chicken B-cells. Vet. Immunol. Immunopathol. 131, 218-28.

Page 115: ENHANCING HOST IMMUNITY TO AVAIAN INFLUENZA VIRUS …

101

Weeratna, R.D., Makinen, S.R., McCluskie, M.J., Davis, H.L., 2005. TLR agonists as

vaccine adjuvants: comparison of CpG ODN and Resiquimod (R-848). Vaccine 23,

5263-70.

Wesche, H., Henzel, W.J., Shillinglaw, W., Li, S., Cao, Z., 1997. MyD88: an adapter that

recruits IRAK to the IL-1 receptor complex. Immunity 7, 837-47.

Wheaton, S., Lambourne, M.D., Sarson, A.J., Brisbin, J.T., Mayameei, A., Sharif, S.,

2007. Molecular cloning and expression analysis of chicken MyD88 and TRIF

genes. DNA Seq. 18, 480-6.

Wong, J.P., Christopher, M.E., Viswanathan, S., Karpoff, N., Dai, X., Das, D., Sun, L.Q.,

Wang, M., Salazar, A.M., 2009. Activation of toll-like receptor signaling pathway

for protection against influenza virus infection. Vaccine 27, 3481-3.

Wong, J.P., Saravolac, E.G., Sabuda, D., Levy, H.B., Kende, M., 1995. Prophylactic and

therapeutic efficacies of poly(IC.LC) against respiratory influenza A virus infection

in mice. Antimicrob. Agents Chemother. 39, 2574-6.