contribution of different components of innate and

298
Contribution of different components of innate and adaptive immunity to severity of flavivirus-induced encephalitis in susceptible and resistant hosts RAFIDAH HANIM SHOMIAD SHUEB (B.SC.) This thesis is presented for the degree of Doctor of Philosophy April 2008 Discipline of Microbiology, School of Biomedical, Biomolecular and Chemical Sciences The University of Western Australia Perth, Western Australia

Upload: others

Post on 23-Mar-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Contribution of different components of innate and

adaptive immunity to severity of flavivirus-induced

encephalitis in susceptible and resistant hosts

RAFIDAH HANIM SHOMIAD SHUEB (B.SC.)

This thesis is presented for the degree of Doctor of Philosophy

April 2008

Discipline of Microbiology, School of Biomedical, Biomolecular and Chemical

Sciences

The University of Western Australia

Perth, Western Australia

STATEMENT

I declare that the work presented in this thesis was conducted by me,

except for intracerebral inoculation which was performed by Dr.

Nadia Urosevic, and the analysis of pathological changes of infected

mouse brains which was done by Prof. Papadimitriou.

………………………………….

Rafidah Hanim Shomiad Shueb

iii

ACKNOWLEDGMENTS

There are people whom I would like to express my deepest gratitude because

without them, this thesis could not be materialised. I will only list their

names here because their immense contribution and assistant is basically

beyond description:

Malaysian Department of Public Services

University of Science Malaysia

Dr. Nadia Urosevic

Dr. Cheryl Johansen

Prof. Geoff Shellam

Prof. John Papadimitriou

Simone Ross

Helen Moulder

Haran, Chris and Lily

Veronica, Giles and Kevin

Juliana and Tobias

Shueb Kaimi (my father) and Zaibah Salim (my mother)

My seven siblings –Anum, Amal, Iqbal, Faiz, Bob, Ali and Ayie

Lazim and my two beautiful kids, Danial and Diyanah.

iv

SUMMARY

Flaviviruses are small, positive-stranded RNA viruses belonging to the family

Flaviviridae. Flavivirus infection in humans could cause diseases ranging from febrile

illnesses to fatal encephalitis. Mice provide a useful small animal model to study

flavivirus-induced encephalitis in humans since mice also develop encephalitis during

flavivirus infection. Some strains of mice have been shown to be resistant to flavivirus

challenge and this resistance is conferred by a single autosomal dominant gene,

designated as Flvr. Recently, OAS1b gene has been identified to be a gene candidate for

Flvr. Several congenic resistant mouse strains have been developed by introducing

resistance genes from outbred or wild mice onto the genetic background of susceptible

C3H mice. These new resistant strains that carry different allelic variants at the Flv

locus include C3H/PRI-Flvr (RV), C3H.MOLD-Flv

mr (MOLD) and C3H.M.domesticus-

Flvr-like (DUB), the latter two being developed in the same laboratory in which the

work described in this thesis was accomplished.

Preliminary studies in this laboratory found that flavivirus resistant mice are vulnerable

to certain flavivirus infections, particularly when challenged by intracerebral (i.c.) route.

Intracerebral (i.c.) challenge with flaviviruses such as West Nile virus (WNV) Sarafend

strain and Kunjin virus (KUNV) MRM16 strain were found to induce high mortality in

flavivirus resistant mice while infection with Murray Valley encephalitis virus (MVEV)

OR2 strain did not cause any apparent disease in the same mice. Based on these

previous findings, this study was designed to further investigate the abrogation of

resistance phenotype expressed in flavivirus resistant DUB mice following infection

with KUNV and to compare a course of infection in resistant versus susceptible mice

with the same virus. Thus, the general aim of this study was to further characterise the

responses of resistant and susceptible mice to KUNV MRM16, to compare these

responses with responses to MVEV OR2 and WNV Sarafend infection and to identify

factors that are associated with a disease development in susceptible and resistant hosts.

Genetic background of the mice and route of virus infection influence the outcome of

infection and because of these, KUNV, MVEV and WNV have different virulence

properties in susceptible and resistant mice. KUNV exhibited low neuroinvasiveness in

adult susceptible and resistant mice but was highly neurovirulent in both strains of mice.

MVEV and WNV were highly neuroinvasive and neurovirulent in susceptible mice.

v

However, only WNV was neurovirulent in resistant mice, while MVEV did not cause

any morbidity to resistant mice at any dose or route of inoculation. Alterations of the

host BBB or depletion of immune cells using different reagents were used in this study

resulting in some changes in the outcome of infection.

To examine the pathogenesis of i.c. infection of KUNV and MVEV, the analysis of

brain histopathology and inflammatory cell infiltrates were performed in both

susceptible HeJ and resistant DUB mice. It was shown in this study that pathogenesis of

KUNV and MVEV was a complex process and the mechanisms involved in susceptible

and resistant mice were different. Several factors including high titres and CD4+ T cells

in the brain contributed to the severe encephalitis observed in KUNV and MVEV-

infected susceptible mice. In contrast, CD8+ T cells had a protective effect in

susceptible HeJ mice MVEV i.c. challenge.

One of the most important findings in this study was that the host immune response

particularly CD8+ T cells and inflammatory mediator, IFNγ, have a strong immuno-

pathological role during lethal i.c. flavivirus infection in the model of flavivirus resistant

mice. This was first time that the immunopathogenic role of T cells in flavivirus

resistant mice was described. The finding is very important as it may provide answer on

the phenomenon seen in the last 50 years regarding the incomplete protection conferred

by the flavivirus resistance gene during certain flavivirus infection in resistant mice.

CD8+ T cells were shown to be the cause of death in resistant DUB mice during i.c.

infection with KUNV as demonstrated by the reduced mortality in mice following T

cells depletion. In contrast, during a non-fatal MVEV infection, T cells were

neuroprotective since the absence of both subsets of T cells, CD4+ and CD8+ T cells,

caused morbidity and mortality in resistant DUB mice infected with MVEV.

Interestingly, T cells were also involved in virus clearance following i.c. infection with

both KUNV and MVEV.

Strong Th1 immune response was induced after KUNV and MVEV i.c. infection in

both susceptible and resistant mice. However, excessive brain IFNγ production at the

time when resistant DUB mice started to exhibit signs of sickness implicated this

cytokine in the development of severe infection of these mice. Using intracellular

labelling of IFNγ, CD8+ T cells were found to be the major producer of this cytokine in

resistant DUB mice. Thus, it can be concluded that CD8+ T cells exerted harmful effect

vi

to resistant DUB mice during KUNV i.c. infection by producing excessive IFNγ that

could be toxic, causing functional loss of the CNS cells.

It was shown from in vitro studies that WNV had the highest tropism for macrophages

and dendritic cells, followed by KUNV. MVEV however did not replicate well in these

cells. This combined with the data from the in vivo studies indicates that macrophages

might be involved in the pathogenesis of intraperitoneal (i.p.) infection of WNV but not

KUNV and MVEV. The reason for this could be that the production of KUNV in

macrophages may not be high enough to induce viraemia and subsequent fatal

encephalitis in mice. In contrast, MVEV appears to use different mechanism or cells for

virus dissemination. Although macrophages may not be involved in KUNV

pathogenesis after i.p. infection, the fact that macrophages support KUNV replication in

vitro may indicate the possibility that blood-borne macrophages were recruited to the

brain where they can get infected with KUNV during i.c. infection and therefore could

participate in KUNV pathogenesis in DUB mice.

This study provides evidence for the first time on the detrimental effect of host antiviral

immunity and inflammatory mediators during flavivirus i.c. infection in resistant mice.

However, it also launches a new question on the selective cell tropism of KUNV versus

MVEV responsible for inducing different pattern of immune responses and

consequently leading to different outcomes of infection in resistant mice.

vii

LIST OF ABBREVIATIONS

ADCC Antibody-dependent cell mediated cytotoxicity

ADE Antibody-dependent enhancement

BAN Banzi virus

BBB Blood brain barrier

BFS Phosphate buffered formalin saline

BRVR Bacteria-resistant-virus-resistant

BRVS Bacteria-resistant-virus-susceptible

C Capsid

oC Degrees Celcius

CaCl2 Calcium chloride

Clodronate Dichloromethylene-biphosphonate

CMI Cell-mediated immunity

cM Centimorgan

CNS Central nervous system

CO2 Carbon dioxide

Con A Concanavalin A

CPE Cytopathic effect

CTL Cytolytic T cells

17D YFV 17D vaccine strain of Yellow fever virus

DAB Diamino methyl benzidine

ddw double distilled water

DEN Dengue

DENV Dengue virus

DHF Dengue haemorrhagic fever

DI Defective interfering

DMSO Dimethyl sulphoxide

DNA Deoxyribonucleic acid

dsRNA double-stranded ribonucleic acid

DTH Delayed-type hypersensitivity

DTT Dithiothreitol

DUB C3H.M.domesticus-Flvr-like

E Envelope

viii

EDTA Ethylenediamine tetra-acetic acid

EMCV Encephalomyocarditis virus

ER Endoplasmic reticulum

FCS Fetal calf serum

g Gram

GAG Glycosaminoglycans

H2O2 Hydrogen peroxide

HA Haemaglutination

HCl Hydrochloric acid

HE Haematoxylin and eosin

He C3H/HeJARC

HeJ C3H/HeJ

HI Haemaglutination inhibition

hr(s) Hour(s)

i.c. intracerebral(ly)

IFN Interferon

IL Interleukin

i.n. intranasal

iNOS inducible nitric oxide synthase

i.p. Intraperitoneal(ly)

I.U. International unit(s)

i.u. infectious unit(s)

i.v. intravenous

JE Japanese encephalitis

JEV Japanese encephalitis virus

kb Kilobases

KCl Potassium chloride

KH2PO4 Potassium dihydrogen orthophosphate

KUN Kunjin

KUNV Kunjin virus

L litre

L929 L929 mouse fibroblast cell line

LPS Lipopolysaccharide

M Molar

MEF Mouse embryo fibroblast

ix

MHC Major histocompatibility class

mg Milligram

mL Millilitre

min Minute(s)

mM Millimolar

MgCl2 Magnesium chloride

MOI Multiplicity of infection

MOLD C3H.MOLD-Flvmr

MRM Mitchell River Mission

mRNA messenger ribonucleic acid

MVE Murray Valley encephalitis

MVEV Murray Valley encephalitis virus

N Neutralising

NaCl Sodium chloride

NaHCO3 Sodium hydrogen carbonate

NaOH Sodium hydroxide

NCR Noncoding region(s)

NCS New born calf serum

NED N-1-napthyethylene diamine dihydrochloride

NGS Normal goat serum

NHS Normal horse serum

NK Natural killer

NO Nitric oxide

NOS Nitric oxide synthase

OAS 2’-5’ oligoadenylate synthetase

OD Optical density

OR Ord River

ORF Open reading frame

PBS Phosphate buffered saline

p.i. Post infection

PRI Princeton-Rockefeller Institute

prM pre-membrane

RER Rough endoplasmic reticulum

RNAse ribonuclease

RF Replicative form

x

RI Replicative intermediate

RNA Ribonucleic acid

rpm rotations per minute

RR Ross river

RSSE Russian spring-summer encephalitis

RT room temperature

RV C3H/PRI-Flvr

rRNA ribosomal ribonucleic acid

s.c. subcutaneous

SDS Sodium dodecyl sulphate

SLEV St. Louis encephalitis virus

ss Single stranded

TBEV Tick-borne encephalitis virus

Tc

Cytotoxic T cells

TCID50 50% tissue culture infectivity dose

TdT Terminal deoxyribonucleotidyl transferase

TMB Tetramethylbenzidine

TNFα Tumor necrosis factor alpha

TUNEL Terminal deoxyribonucleotidyl transferase dUTP nick end

labeling

µg Microgram

µL Microlitre

US United States

UTR Untranslated region

UWA University of Western Australia

Vero African Green Monkey kidney

WN West Nile

WNV West Nile virus

YF Yellow Fever

YFV Yellow fever virus

xi

TABLE OF CONTENTS

1.0 CHAPTER 1: LITERATURE REVIEW ------------------------------ 1

1.1 FLAVIVIRUS IN GENERAL --------------------------------------------------------- 1

1.1.1 HISTORY AND CLASSIFICATION ------------------------------------------ 1

1.1.2 ECOLOGY AND EPIDEMIOLOGY------------------------------------------- 2

1.1.3 MORPHOLOGY ------------------------------------------------------------------- 4

1.1.5 VIRAL PROTEINS --------------------------------------------------------------- 5

1.1.5.1 Structural proteins ----------------------------------------------------------- 6

1.1.5.2 Non-structural proteins ----------------------------------------------------- 7

1.1.6 VIRUS ENTRY AND TRANSLATION --------------------------------------- 8

1.1.7 REPLICATION -------------------------------------------------------------------- 9

1.1.8 VIRUS ASSEMBLY AND RELEASE ---------------------------------------- 10

1.2 PATHOGENESIS OF FLAVIVIRUSES ------------------------------------------ 11

1.2.1 INFECTION IN VERTEBRATE HOSTS ------------------------------------ 12

1.2.2 UP-REGULATION OF HOST CELL SURFACE MOLECULES

UPON FLAVIVIRUS INFECTION ------------------------------------------- 13

1.2.3.1 Nature and properties of the central nervous system ------------------ 14

1.2.3.2 Flavivirus infections in central nervous system------------------------ 17

1.2.4 NEUROINVASIVENESS AND NEUROVIRULENCE ------------------- 18

1.3 IMMUNE RESPONSE AND IMMUNOPATHOLOGY ---------------------- 20

1.3.1 INNATE IMMUNE SYSTEM -------------------------------------------------- 21

1.3.1.1 Macrophages ---------------------------------------------------------------- 21

1.3.1.2 Nitric oxide ------------------------------------------------------------------ 22

1.3.1.3 Natural killer cells ---------------------------------------------------------- 23

xii

1.3.1.4 Neutrophils ------------------------------------------------------------------ 23

1.3.2 ADAPTIVE IMMUNITY ------------------------------------------------------- 24

1.3.2.1 Humoral mediated immunity --------------------------------------------- 25

1.3.2.2 Cell-mediated immunity -------------------------------------------------- 27

1.3.3 SOLUBLE MEDIATORS ------------------------------------------------------- 30

1.4.3.1 Cytokines -------------------------------------------------------------------- 30

1.3.3.1.1 IFN type I ------------------------------------------------------------- 31

1.3.3.1.2 IFNγ ------------------------------------------------------------------- 33

1.3.3.1.3 TNF ------------------------------------------------------------------ 34

1.3.3.2 Chemokines ----------------------------------------------------------------- 35

1.4 GENETIC RESISTANCE TO FLAVIVIRUSES ------------------------------- 36

1.4.1 FLAVIVIRUS RESISTANCE IN HUMANS -------------------------------- 37

1.4.2 FLAVIVIRUS RESISTANCE IN MURINE MODELS -------------------- 37

1.4.2.1 History and development ------------------------------------------------- 37

1.4.2.2 Flavivirus resistance in wild mice --------------------------------------- 38

1.4.2.3 Development of congenic flavivirus mouse resistant strains -------- 39

1.4.2.4 Resistance expression in mice -------------------------------------------- 40

1.4.2.5 Resistance expression in cell culture ------------------------------------ 40

1.4.3 THE MECHANISM OF FLAVIVIRUS RESISTANCE ------------------- 41

1.4.4 ANALYSIS OF GENE CANDIDATE FOR FLAVIVIRUS

RESISTANCE GENE ------------------------------------------------------------ 43

1.4.5 FACTORS INFLUENCING THE HOST INNATE RESISTANCE TO

FLAVIVIRUSES ----------------------------------------------------------------- 44

1.5 AIMS -------------------------------------------------------------------------------------- 46

xiii

2.0 CHAPTER 2: MATERIALS -------------------------------------------- 49

2.1. REAGENTS ---------------------------------------------------------------------------- 49

2.2 CELL CULTURE MATERIALS --------------------------------------------------- 52

2.3 BUFFERS, SOLUTIONS AND MEDIA ------------------------------------------- 53

2.3.1 CELL STUDIES ------------------------------------------------------------------ 53

2.3.1.1 Growth media --------------------------------------------------------------- 53

2.3.1.2 Cell culture solutions ------------------------------------------------------ 54

2.3.2 IMMUNOHISTOCHEMISTRY ------------------------------------------------ 55

2.3.3 FLOW CYTOMETRY ----------------------------------------------------------- 57

2.3.4 CELL ISOLATION -------------------------------------------------------------- 57

2.3.5 ELISA REAGENTS -------------------------------------------------------------- 58

3.0 CHAPTER 3: METHODS ----------------------------------------------- 59

3.1 VIRUSES --------------------------------------------------------------------------------- 59

3.1.1 VIRUS STRAINS ---------------------------------------------------------------- 59

3.1.2 PROPAGATION OF VIRUS STOCKS --------------------------------------- 59

3.2 ANIMAL STUDIES -------------------------------------------------------------------- 60

3.2.1 MOUSE STRAINS --------------------------------------------------------------- 60

3.2.2 VIRUS INOCULATION OF MICE ------------------------------------------- 60

3.2.2.1 Intracerebral inoculation -------------------------------------------------- 60

3.2.2.2 Intraperitoneal inoculation ------------------------------------------------ 61

3.2.2.3 Intranasal inoculation ------------------------------------------------------ 61

xiv

3.3 INOCULATION OF REAGENTS/CELLS INTO MICE ---------------------- 61

3.3.1 THIOGLYCOLLATE ------------------------------------------------------------ 61

3.3.2 LIPOPOLYSACCHARIDE (LPS) --------------------------------------------- 62

3.3.3 SODIUM DODECYL SULPHATE (SDS) ---------------------------------- 62

3.3.4 CD4+ AND CD8 T+ CELLS DEPLETION ---------------------------------- 62

3.4.5 CLODRONATE ------------------------------------------------------------------ 63

3.4 ORGAN EXTRACTION -------------------------------------------------------------- 63

3.4.1 BRAINS ---------------------------------------------------------------------------- 64

3.4.2 PERIPHERAL ORGANS ------------------------------------------------------- 64

3.5 CELL ISOLATION -------------------------------------------------------------------- 65

3.5.1 BRAIN MONONUCLEAR CELLS ------------------------------------------- 65

3.5.2 SPLENOCYTES ------------------------------------------------------------------ 65

3.5.3 PERITONEAL MACROPHAGES --------------------------------------------- 66

3.5.3.1 In vitro experiments -------------------------------------------------------- 66

3.5.3.2 In vivo experiments -------------------------------------------------------- 67

3.6 HISTOLOGICAL PREPARATION AND

IMMUNOHISTOCHEMISTRY OF ORGANS --------------------------------- 68

3.6.1 BRAIN ------------------------------------------------------------------------------ 68

3.6.1.1 Paraffin embedding of the brain ----------------------------------------- 68

3.6.1.2 Hematoxylin and eosin (HE) staining ----------------------------------- 68

3.6.1.3 Activated brain microglia/macrophages labeling ---------------------- 69

3.6.1.4 Detection of macrophages in spleens ------------------------------------ 69

3.6.1.4.1 Cryosectioning of spleen ------------------------------------------- 69

3.6.1.4.2 Detection of macrophages ------------------------------------------ 70

xv

3.6.1.5 Apoptosis Detection ------------------------------------------------------- 70

3.7 CELL STUDIES ------------------------------------------------------------------------ 71

3.7.1 AFRICAN GREEN MONKEY KIDNEY CELLS (VERO CELLS)

AND L292 MOUSE FIBROBLASTS ----------------------------------------- 71

3.7.2 HYBRIDOMA YTS 191 AND 169 CELL LINES -------------------------- 71

3.7.2.1 Cell culture ------------------------------------------------------------------ 71

3.7.2.2 Production of anti CD4+ and anti CD8+ antibodies ------------------ 72

3.7.2.3 Ammonium sulfate precipitation ----------------------------------------- 72

3.7. 3 MOUSE PRIMARY CELL CULTURES ------------------------------------ 73

3.7.4 VIRUS INFECTION OF CELLS ---------------------------------------------- 73

3.7.6 VIRUS TITRATION ------------------------------------------------------------- 74

3.7.6.1 Preparation of 10% brain homogenates --------------------------------- 74

3.7.6.2 Tissue culture infectivity dose 50% (TCID50) ------------------------- 74

3.8 CYTOKINE STUDIES ---------------------------------------------------------------- 75

3.8.1 IFN TYPE I BIOASSAY -------------------------------------------------------- 75

3.8.1.1 Preparation of L929 monolayers ----------------------------------------- 75

3.8.1.2 Acid treatment of samples ------------------------------------------------ 76

3.8.1.3 IFN type I bioassay -------------------------------------------------------- 76

3.9 FLOW CYTOMETRY ---------------------------------------------------------------- 77

4.0 CHAPTER 4: STUDY ON KUNV, MVEV AND WNV

VIRULENCE IN SUSCEPTIBLE AND CONGENIC

RESISTANT MICE ------------------------------------------------------- 80

4.1 INTRODUCTION ---------------------------------------------------------------------- 80

xvi

4.2 RESULTS -------------------------------------------------------------------------------- 81

4.2.1 VIRUS NEUROVURULENCE STUDIES ----------------------------------- 81

4.2.1.1 Analysis of neurovirulence of WNV, KUNV and MVEV in

susceptible mice ------------------------------------------------------------ 81

4.2.1.2 Analysis of neurovirulence of WNV, KUNV and MVEV in

resistant mice --------------------------------------------------------------- 85

4.2.1.3 Analysis of different degrees of neurovirulence of WNV,

KUNV and MVEV --------------------------------------------------------- 85

4.2.1.3 Mouse mortality and average time to death using a 100 LD50

virus dose -------------------------------------------------------------------- 87

4.2.2 INTRANASAL INFECTION IN SUSCEPTIBLE MICE ------------------ 88

4.2.3 VIRUS NEUROINVASIVENESS STUDIES -------------------------------- 91

4.2.3.1 Intraperitoneal challenge in adult and young mice -------------------- 91

4.2.3.2 Effect of blood brain barrier modulation on virus

neuroinvasiveness ---------------------------------------------------------- 94

4.2.3.2.1 Effect of SDS on KUNV and MVEV neuroinvasiveness in

HeJ mice -------------------------------------------------------------- 94

4.2.3.2.2 Effect of blood brain barrier modulation on WNV

neuroinvasiveness in mice ----------------------------------------- 96

4.2.3.3.1 Mouse survival following thioglycollate treatment ------------ 99

4.2.3.3.2 Mouse survival following transient macrophage depletion - 102

4.2.3.4 Effect of T cells depletion on survival of DUB mice following

WNV i.p. infection ------------------------------------------------------- 106

4.3 DISCUSSION ------------------------------------------------------------------------- 109

xvii

5.0 CHAPTER 5: CHARACTERISATION OF KUNV, MVEV

AND WNV INFECTIONS IN CELL CULTURE ----------------- 118

5.1 INTRODUCTION -------------------------------------------------------------------- 118

5.2 RESULTS ------------------------------------------------------------------------------ 119

5.2.1 VIRUS REPLICATION IN CELL CULTURE ---------------------------- 119

5.2.1.1 Determination of dose of infection ------------------------------------ 119

5.2.1.2 Virus replication in Vero cells ----------------------------------------- 119

5.2.1.3 Virus repl

ication in thioglycollate-elicited macrophages -------------------------------- 120

5.2.1.4 Virus replication in primary mouse dendritic cells ------------------ 122

5.2.2 CYTOKINE PRODUCTION IN PRIMARY MOUSE

MACROPHAGES -------------------------------------------------------------- 129

5.2.3 ADOPTIVE TRANSFER OF VIRUS-INFECTED MACROPHAGES

IN MICE ------------------------------------------------------------------------- 133

5.3 DISCUSSION ------------------------------------------------------------------------- 137

6.0 CHAPTER 6: ROLE OF VIRAL REPLICATION AND

IMMUNOPATHOLOGY IN DISEASE DEVELOPMENT

FOLLOWING KUNV AND MVEV INTRACEREBRAL

INFECTION --------------------------------------------------------------- 142

6.1 INTRODUCTION -------------------------------------------------------------------- 142

6.2 RESULTS ------------------------------------------------------------------------------ 144

xviii

6.2.1 BRAIN VIRUS TITRES FOLLOWING INTRACEREBRAL

INFECTION --------------------------------------------------------------------- 144

6.2.1.2 Analysis of viral titres in mouse brain following KUNV and

MVEV infection ---------------------------------------------------------- 144

6.2.1.2 Analysis of viral titres in peripheral organs following KUNV

and MVEV infection ----------------------------------------------------- 147

6.2.2 BRAIN HISTOPATHOLOGICAL AND INFLAMMATION

ANALYSIS ---------------------------------------------------------------------- 149

6.2.2.1 Brain architecture and inflammation in KUNV and MVEV

infection ------------------------------------------------------------------- 149

6.2.2.2 Brain tissue architecture and leucocytic infiltration in the brains

of infected mice ---------------------------------------------------------- 149

6.2.2.3 Analysis of accumulation and activation of

microglia/macrophages in the brains of virus-infected mice ------ 157

6.2.2.4 Contribution of apoptosis to fatal outcome of infection ------------ 158

6.3 DISCUSSION ------------------------------------------------------------------------- 165

7.0 CHAPTER 7: ROLE OF CELL MEDIATED IMMUNITY IN

IMMUNOPATHOLOGY OR RECOVERY FOLLOWING

INTRACEREBRAL KUNV AND MVEV INFECTION IN

MICE ----------------------------------------------------------------------- 172

7.1 INTRODUCTION -------------------------------------------------------------------- 172

7.2 RESULTS ------------------------------------------------------------------------------ 174

7.2.1 FLOW CYTOMETRIC ANALYSIS OF BRAIN MONONUCLEAR

CELLS FOLLOWING KUNV AND MVEV INFECTION -------------- 174

xix

7.2.1.1 Analysis of cells infiltrating the brains of susceptible HeJ mice

upon infection with MVEV and KUNV ------------------------------ 174

7.2.1.3 Analysis of MHC cell surface up-regulation on brain CD11b+

cells following flavivirus infection. ----------------------------------- 186

7.2.2 T CELL DEPLETION STUDIES -------------------------------------------- 188

7.2.2.1 Pilot study to determine the optimum antibody depletion time --- 190

7.2.2.2 Effect of CD4+ or CD8+ T cells depletion on mortality

following flavivirus infection in susceptible mice ------------------ 192

7.2.2.3 Effect of CD4+ or CD8+ T cells depletion on mortality

following flavivirus infection in resistant DUB mice --------------- 193

7.2.2.4 Effect of total T cells (CD4+ and CD8+) depletion on mortality

following KUNV and MVEV infection in resistant DUB mice --- 197

7.2.3 ANALYSIS OF CYTOKINE PRODUCTIONS --------------------------- 202

7.2.3.1 Cytokine productions in susceptible HeJ mice----------------------- 203

7.2.3.2 Cytokine productions in resistant DUB mice ------------------------ 205

7.2.3.3 Analysis of major IFNγ producing cells in resistant DUB mice -- 208

7.3 DISCUSSION ------------------------------------------------------------------------- 213

8.0 CHAPTER 8: GENERAL DISCUSSION --------------------------- 223

9.0 REFERENCES ----------------------------------------------------------- 236

xx

TABLE OF FIGURES

Figure 4.1. Analysis of survival in mice following infection with 100

LD50 (in susceptible mice) of KUNV and MVEV.

8

Figure 4.2. Depletion of splenic macrophages by clodronate treatment. 105

Figure 5.1. Replication of WNV, KUNV and MVEV in Vero cells. 123

Figure 5.2. Replication of WNV, KUNV and MVEV in A)

thioglycollate-elicited macrophages from flavivirus

susceptible HeJ mice and B) resistant DUB mice.

124

Figure 5.3. Replication of WNV, KUNV and MVEV in C57/BL6 mouse

bone marrow derived dendritic cells.

126

Figure 5.4. Cytopathic effect of virus replication in Vero cells. 127

Figure 5.5. Cytopathic effect of virus replication in macrophage cell

cultures.

128

Figure 5.6. In vitro cytokine productions by HeJ isolated macrophages

following infection with WNV, KUNV and MVEV.

131

Figure 6.1. Kinetics of viral replication in mouse brains infected with

KUNV and MVEV.

148

Figure 6.2. Brain tissue section from uninfected mouse. 153

Figure 6.3. Brain tissue inflammation in susceptible HeJ mice infected

with KUNV (A and B) or MVEV (C and D) at the time of

death.

154

Figure 6.4. Brain tissue inflammation on day 5 p.i. in DUB mice

infected with KUNV (A and B) or MVEV (C and D).

155

Figure 6.5. Brain tissue inflammation on day 9 p.i. following i.c. KUNV

(A and B) and MVEV (C and D) infection in DUB mice.

156

Figure 6.6. Detection of activated microglia/macrophages in the brains

of susceptible mice following i.c. KUNV and MVEV

infection.

160

Figure 6.7. Analysis of apoptosis in brains of susceptible and resistant

mice following i.c. KUNV and MVEV infection.

163

Figure 7.1. Total number of cells isolated from (A) spleens and (B)

brains of resistant mice challenged i.c. either with KUNV or

178

xxi

MVEV.

Figure 7.2. Flow cytometric analysis of splenocytes in DUB mice

following i.c. KUNV and MVEV infection.

180

Figure 7.3. Analysis of brain infiltrating leucocytes in DUB mice

following KUNV and MVEV infection.

183

Figure 7.4. Up-regulation of MHC class I and II molecules in CD11b+

cells following KUNV and MVEV infection.

189

Figure 7.5. Analysis of T cells depletion in DUB mice by flow

cytometry after treatment with cytotoxic anti-CD4 and anti-

CD8 antibodies.

191

Figure 7.6. Effect of CD4 or CD8 cells depletion on mortality following

KUNV and MVEV infection in flavivirus susceptible HeJ

mice.

194

Figure 7.7. Effect of CD4+ or CD8+ T cells depletion on mortality of

resistant DUB mice following challenge with KUNV and

MVEV.

198

Figure 7.8. Effect of CD4+ or CD8+ T cells depletion on viral titres

following MVEV infection in resistant DUB mice.

198

Figure 7.9. Effect of T cells (CD4+ and CD8+) depletion on mortality of

KUNV and MVEV-infected resistant DUB mice.

199

Figure 7.10. Brain IFNαβ (A) and TNF (B) levels in resistant DUB mice

following infection with KUNV and MVEV.

207

Figure 7.11. Analysis of IFN producing cells in resistant DUB mouse

brains 7 days after infection with KUNV or MVEV.

212

xxii

TABLE OF TABLES

Table 1.1. Recent classification of the Flavivirus genus 3

Table 4.1.A. Mortality and LD50 studies following intracerebral infection

with serially diluted viruses in flavivirus susceptible HeJ

mice.

83

Table 4.1.B. Mortality in different susceptible mouse strains following

intracerebral infection with KUNV.

84

Table 4.1.C. Mortality and LD50 studies following intracerebral infection

with serially diluted viruses in flavivirus resistant DUB mice.

86

Table 4.2. Mortality studies following intranasal infection of KUNV

and MVEV in HeJ mice.

90

Table 4.3 Intraperitoneal infection of KUNV, MVEV and WNV in

flavivirus susceptible HeJ and resistant DUB mice.

93

Table 4.4. The effect of SDS on mortality of HeJ mice following i.p.

KUNV and MVEV infection.

98

Table 4.5. The effect of SDS on mortality of mice following i.p. WNV

infection in mice.

101

Table 4.6. The effect of thioglycollate on mortality of HeJ and DUB

mice following i.p. virus infection.

103

Table 4.7. The effect of macrophage and blood brain barrier modulation

on WNV and KUNV infections in young DUB mice.

104

Table 4.8. The effect of T cells depletion on mortality of DUB mice

following i.p. WNV infection

108

Table 5.1. Mortality studies following i.p. infection of mice with HeJ

peritoneal macrophages infected in vitro with WNV.

134

Table 5.2. Mortality studies following i.p. infection of mice with DUB

peritoneal macrophages infected in vitro with WNV.

135

Table 6.1. The KUNV and MVEV doses used for intracerebral

infection in mice

145

Table 6.2. Analysis of viral titres in peripheral organs of KUNV or

MVEV-infected HeJ mice at the time of death.

150

Table 6.3. Summary of viral titres, histopathology and microglia 162

xxiii

analysis in susceptible and resistant mice following i.c.

infection with KUNV and MVEV.

Table 7.1. Number of brain infiltrating leucocytes isolated from HeJ

mice that succumbed to KUNV and MVEV infection.

177

Table 7.2. Effect of CD4+ and CD8+ T cells depletion on mortality of

HeJ mice following i.c. challenge with KUNV or MVEV.

194

Table 7.3. Summary on the effect of T cells depletion in DUB mice

challenged with KUNV or MVEV.

200

Table 7.4. Cytokine levels in brains of susceptible HeJ mice at the time

of death following infection with KUNV or MVEV.

204

Table 7.5. Th1-Th2 cytokines in DUB mouse sera following infection

with KUNV and MVEV.

210

Table 7.6. Th1-Th2 cytokines in DUB mouse brains following infection

with KUNV and MVEV.

211

CHAPTER 1

LITERATURE REVIEW

1

1.0 CHAPTER 1: LITERATURE REVIEW

1.1 FLAVIVIRUS IN GENERAL

1.1.1 HISTORY AND CLASSIFICATION

Yellow fever virus (YFV) was the first mosquito-transmitted virus known to cause

diseases in humans. It was also the first arbovirus to be isolated (1927) and cultivated

(1932) (reviewed in Burke and Monath, 2001). When a number of other arboviruses

were also discovered, the Togaviridae family was established to group these viruses

together. Two groups of viruses were classified within this family, known as Group A

and Group B, which shared similarity in modes of transmission, viral genome

organisation and morphology but were serologically distinct. However, further studies

revealed that there were significant differences between these two groups of viruses in

terms of viral replication, genome structure and gene order. Thus, Group A was later

reclassified as Alphavirus genus in the family Togaviridae whereas group B was known

as Flavivirus genus (from Latin word Flavus which means yellow, for yellow fever) and

incorporated into a new virus family known as Flaviviridae (Westaway et al, 1985;

Westaway, 1987).

At present, in addition to the genus Flavivirus, the family Flaviviridae also includes

another 2 genera; Pestivirus (from the Latin word pestis which means plague) and

Hepacivirus (from Greek word hepatos which means liver). These 3 groups of viruses

have similar morphology, genome structures and replication strategies (Heinz et al,

2000). However, Pestivirus and Hepacivirus are not arthropod-borne and they do not

share any antigenic resemblance with Flavivirus (Tsai, 2000). The Flavivirus genus

consists of approximately 70 members, which are further classified into 10 subgroups or

complexes (Table 1.1) (ICTVdB management, 2006), based upon cross-neutralisation in

plaque reduction neutralisation test and vector species involved in transmission

(reviewed in Calisher and Gould, 2003). Classifying the viruses according to their

vectors nevertheless is not always appropriate as some mosquito-borne flaviviruses have

also been isolated from ticks and vice versa. Hence, advanced computer programs and

molecular biological techniques provides excellent tools for extensive nucleotide

CHAPTER 1

LITERATURE REVIEW

2

sequence comparisons, which subsequently make it possible to draw a more accurate

genetic relationship and taxonomy of flaviviruses (Calisher and Gould, 2003).

Most flaviviruses are arboviruses; they are either transmitted by ticks or mosquitoes.

Currently there are 31 mosquitoes-borne Flavivirus species, 12 tick-borne species and

14 species with no known vector (ICTVdB management, 2006).

1.1.2 ECOLOGY AND EPIDEMIOLOGY

Arboviruses including flaviviruses are globally distributed. Among the 10 subgroups of

flaviviruses, Japanese encephalitis (JE) serocomplex contains the most medically

important viruses that are found in all continents except in the Antartic (Mackenzie et

al, 2002a). Natural transmission cycles of these viruses are primarily maintained

between arthropod vectors and susceptible vertebrate hosts like birds (Mackenzie et al,

2002a). Horse and human infections with flaviviruses are considered incidental except

in the case of dengue (DEN) and yellow fever (YF) infections where humans may also

be involved in virus transmission (Vasconcelos et al, 2001; Ligon, 2005).

WNV is the most widely distributed member of the JE complex and can be found in

Africa, the Middle East, Europe, Asia, North America and Australia (Mackenzie et al,

2004). Currently, West Nile virus is divided into lineage I and II. The Lineage I includes

viruses isolated from Africa, Europe, Asia, North America and Australia and commonly

associated with human diseases (Lanciotti et al, 1999; Mackenzie et al, 2004). Lineage

II consists solely of viruses isolated from southern Africa and Madagascar (Lanciotti et

al, 1999). Previously, human infection with WNV resulted mostly in fever-arthralgia-

rash syndrome but recent WNV outbreaks in Europe and The United States involving

large numbers of encephalitic cases has sparked serious concerns about the re-

emergence of WNV with high virulence (Solomon and Vaughn, 2002). In 2002, a large

outbreak of WNV occurred in North America with more that 4000 people were infected

and 250 fatalities were recorded (Lanciotti et al, 2002).

CHAPTER 1

LITERATURE REVIEW

3

Table 1.1 Recent classification of the Flavivirus genus (ICTVbB management,

2006)

Group Vector Viruses

Mammalian tick-

borne

encephalitis

Tick

Gadgets Gully, Kyasanur Forest disease,

Langat, Louping ill, Omsk hemorrhagic

fever, Powassan, Royal Farm, Tick-borne

encephalitis

Seabird tick-

borne

encephalitis

Tick Kadam, Meaban, Saumarez Reef, Tyuleniy

Aroa Virus Mosquito Aroa

Yellow Fever Mosquito

Banzi, Bouboui, Edge Hill, Jugra, Saboya,

Sepik, Uganda S, Wesselsbron, Yellow

fever

Japanese

encephalitis Mosquito

Japanese encephalitis, St. Louis

encephalitis, Murray Valley encephalitis,

West Nile, Kunjin, Kokobera, Usuta,

Stratford, Alfuy, Koutango

Dengue Mosquito Dengue-1, Dengue-2, Dengue-3, Dengue-4

Kokobera Mosquito Kokobera

Ntaya Mosquito

Bagaza, Ilheus, Israel turkey

meningoencephalomyelitis, Ntaya,

Tembusu

Spondweni Mosquito Zika

Entebbe bat

No known

arthropod

vector

Entebbe bat, Yokose, Apoi, Cowbone

Ridge, Jutiapa, Modoc, Sal Vieja virus, San

Perlita, Bukalasa bat, Carey Island, Dakar

bat, Montana myotis leukoencephalitis,

Phnom Penh bat, Rio Bravo

CHAPTER 1

LITERATURE REVIEW

4

Later, sequence analysis indicated that the WNV strain responsible for the North

America outbreak was closely related to a WNV strain that was isolated from Israel

during 1998-2000 outbreaks (Lanciotti et al, 2002).

In Australia, KUNV, an Australian variant of WNV, can be found. This virus has a

similar distribution to the major Australian encephalitogenic flavivirus, MVEV, and

both are endemic in several areas including the northern part of Western Australia,

Northern Territory and northern Queensland (Hall et al, 2002). These two viruses share

similar ecologies, as they have the same vector and vertebrate hosts (reviewed in

Mackenzie et al, 2002b). Although KUNV isolates from Australia belong to the lineage

I of WNV, they are only associated with mild febrile illness in humans (Mackenzie et

al, 2004; Hall et al, 2002). In addition to Australia, KUNV also can be found in Papua

New Guinea, Irian Jaya, Indonesian archipelago as well as some parts of Southeast Asia

(reviewed in Hall et al, 2002). However, genetic analysis indicates that KUNV isolates

from Southeast Asia may have evolved separately from a common ancestor and thus are

a separate lineage of WNV (Scherret et al, 2001).

MVEV is the major encephalitogenic flavivirus in Australia that was first isolated from

humans in 1951 during a major outbreak in south-eastern Australia. MVEV was also

isolated from Culex annulirostris mosquitoes in 1959 at Kowanyama, Cape York,

northern Queensland (reviewed in Mackenzie et al, 2002a).

1.1.3 MORPHOLOGY

Virions of flaviviruses are spherical, approximately 40-60 nm in diameter. They have an

icosahedral core 30-35 nm in diameter, which contains a nucleocapsid protein

complexed with RNA (Duane and Roehrig, 2000). The nucleocapsid core is surrounded

by a lipid bilayer envelope, which is densely covered with surface projections,

consisting of membrane (M) and envelope (E) proteins (Duane and Roehrig, 2000).

Both M and E proteins are anchored to the envelope by hydrophobic bonds (Burke and

Monath, 2001).

CHAPTER 1

LITERATURE REVIEW

5

1.1.4 GENOME STRUCTURE

The genome of flaviviruses consists of a linear positive single stranded RNA of about

10.4 to 11kb in length (Brinton, 1986). The molecular weight is estimated to be about

4 x 103 kDA (Westaway et al, 1985). The 5’ end has a type 1 cap followed by the

conserved dinucleotide sequence AG while the 3’ end has no polyA tail (Duane and

Roehrig, 2000; Chambers et al, 1990a).

There is only a single open reading frame (ORF) of more than 10,000 bases which is

flanked by 5’ (95 to 132 bases) and 3’ short un-translated regions (UTR) (114 to 624

bases) that contain conserved RNA elements (Rice, 1996). The 5’ non-translated region

is believed to carry stable secondary structure involved in recognition and binding of

viral and host replication and packaging factors (Lindenbach and Rice, 2001). Most

flaviviruses also carry a 3’ terminal stem-loop structure at the 3’ non-coding region

which may function as a promoter to initiate negative strand RNA synthesis. The 3’

stem-loop structure has been shown to bind to viral helicase NS3 and NS5 in vitro

(reviewed in Lindenbach and Rice, 2003). Upstream of the 3’ stem loop structure is

another RNA element known as cyclisation sequence (CS) which is complementary to a

sequence present at the 5’ end of the genome and is necessary for replication

(Khromykh et al, 2001; Alvarez et al, 2005). The ORF encodes and directs synthesis of

a single polyprotein that serves as a precursor to a number of viral proteins. The

ORF is arranged as follows: C-prM-NS1-NS2A-NS2B-NS3-NS4A-NS4B-NS5 (Rice

et al., 1985). Complete genome sequences are already known for several flaviviruses

including YFV (Rice et al, 1985), WNV (Castle et al, 1985), Japanese encephalitis virus

(JEV) (Sumiyoshi et al, 1987), MVEV (Dalgarno et al, 1986) and KUNV (Coia et al,

1988).

1.1.5 VIRAL PROTEINS

Translation results in the synthesis of a single polyprotein, which eventually cleaved by

host and viral proteases into 10 proteins (Chambers et al, 1990a). Of this, 3 are

structural; capsid (C), pre-membrane (prM) and envelope (E) and 7 are non-structural

proteins; NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5.

CHAPTER 1

LITERATURE REVIEW

6

1.1.5.1 Structural proteins

The C protein is a small but highly basic protein. The basic residues are mainly at the N

and C termini of the protein structure and probably act in concert to bind viral RNA. A

hydrophobic domain at the central part of the C protein interacts with cellular

membranes and is involved in virion morphogenesis (Lindenbach and Rice, 2001). The

C protein is part of the component of nucleocapsid core structure and contributes to the

antigenic group reactivity as detected by the complement-fixation test (Rice 1996;

Duane and Roehrig, 2000). The nascent C protein contains a C-terminal hydrophobic

anchor that provides the signal for ER translocation of prM (Lindenbach and Rice,

2003).

The prM protein is a glycosylated intracellular precursor of the virion associated M

protein (Heinz et al, 2000). PrM and E proteins form a heterodimeric complex shortly

after synthesis and prM serves as a chaperone to E protein within the cell secretory

pathways to prevent it from misfolding (Tsai, 2000). The maturation process of virions

in the secretory pathway takes place concurrently with the cleavage of prM into pr and

M proteins by host furin enzyme (Murray et al, 1993). Following cleavage, M protein is

found in mature virions while the pr portion is secreted independently (Murray et al,

1993). PrM proteins are capable of eliciting protective neutralising antibodies

(Lindenbach and Rice, 2001).

E protein is involved in many biological processes including virion assembly, cell

receptor recognition, fusion with cell endosomal membranes, agglutination of red blood

cells, viral tropism and pathogenesis (Deubel et al, 2001; Hurrelbrink and McMinn,

2001). In addition, the E protein is also associated with the generation of neutralising,

enhancing and protective antibodies (Burke and Monath, 2001). In some viruses like

WNV, the E protein is N-glycoslyated and this structural modification may change viral

neuroinvasiveness and neurovirulence (Halevy et al, 1994; Chambers et al, 1998).

Three antigenic domains have been identified in the E protein so far; I, II and III (Mandl

et al, 1989; Heinz and Roehrig, 1990). Domain I is believed to contain most of the virus

conformational virus antigenic epitopes while domain III has been proposed to contain

putative receptor-binding region (Duane and Roehrig, 2000)

CHAPTER 1

LITERATURE REVIEW

7

1.1.5.2 Non-structural proteins

Unlike the structural proteins, functions of non-structural proteins are not fully

understood. NS1, NS3 and NS5 are large and highly conserved proteins while the rest

are small hydrophobic and poorly conserved non-structural proteins (Chambers et al,

1990a).

NS1 is a membrane-bound glycoprotein but it can also be secreted from virus infected-

cells. This protein is likely to be involved in virion morphogenesis as well as viral RNA

synthesis (Duane and Roehrig, 2000; Mackenzie et al, 1996). Mutations at the N-linked

glycosylation sites of this protein affect RNA and virus production (Muylaert et al,

1996). A strong humoral immune response has also been documented against the

secreted form of NS1 while the membrane-bound form of this protein can induce

antibodies that direct the complement-mediated lysis of virus-infected cells (reviewed in

Lindenbach and Rice, 2003). On the contrary, recently, a possible role of NS1 in

attenuating complement activation during certain flavivirus infections has been

suggested. Chung and co-workers (2006) demonstrated that WNV NS1 binds to and

recruits factor H (fH), which is a key regulatory molecule of complement activation.

This activity results in the inhibition of complement-mediated immunity of infected

hosts (Chung et al, 2006)

NS3 is a multifunctional protein and serves as a viral helicase during viral RNA

replication while NS2B-NS3 complex functions as a viral protease associated with

cleavage of viral polyprotein (Yamshchikov and Compans, 1993; Chambers et al,

1990b, Wu et al, 2005). NS4A and NS4B have molecular weights of 16 and 27 kDa,

respectively. Their function is not clear but it is possible that these proteins have a role

in RNA replication (Lindenbach and Rice, 1999). NS5 has a molecular weight of 103 kb

and thus is the largest and the most conserved flavivirus protein (Lindenbach and Rice,

2001). Given the high level of sequence homology to RNA-dependent RNA

polymerases (RdRps) of other positive strand RNA viruses, NS5 may serve as a viral

polymerase (Lindenbach and Rice, 2001; Chambers et al, 1990a). In addition, the NS5

protein might also function as a methyltransferase enzyme, engaging in the methylation

of the 5’ RNA cap structure (Lindenbach and Rice, 2001). NS5 has also been shown to

form a complex with NS3 resulting in NS3 NTPase activity (Lindenbach and Rice,

2001).

CHAPTER 1

LITERATURE REVIEW

8

1.1.6 VIRUS ENTRY AND TRANSLATION

Flaviviruses are capable of infecting a wide range of invertebrate and vertebrate hosts,

as well as mammalian, avian and arthropod cell lines. Initial attachment of the virion is

mediated by the E protein, which binds to the host cell surface receptor. At present, this

cell surface receptor has not been fully identified and characterised either in the

periphery or central nervous system (CNS) but the availability of these receptors

determines tissue and cell tropism as well as the host range of the virus. Recently,

CD209 or DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing

nonintegrin) has been suggested as a cell surface receptor for dengue virus (DENV)

(reviewed in Chambers and Diamond, 2003). However, the significance of this receptor

in vivo is unknown and further study is required to determine whether other flaviviruses

utilise the same ligand for attachment or entry (Chambers and Diamond, 2003). Highly

sulfated glycosaminoglycans were shown to promote the binding of E protein and

heparin sulfate expression on the host cell surface was required for efficient infection

with laboratory-passaged DENV and MVEV strains (Chen et al, 1997; Lee and Lobigs,

2002). However the role of heparin as an important cell surface receptor to mediate

virus entry remains uncertain as serial laboratory passages of JEV and MVEV showed

increased binding for this receptor in vitro but at the same time the viruses had low

virulence in vivo (Lee and Lobigs, 2002). In vitro analysis of WNV Sarafend strain

infection revealed that the host αVβ3 intergrin served as a functional receptor that

interacted with domain III of the envelope protein to mediate virus binding (Lee et al,

2006). In addition, binding may also occur through antibody-dependent enhancement

(ADE) as has been shown in DENV infection. In this instance, virus particles are

opsonised with subneutralising concentrations of antibodies and bound to cells

expressing immunoglobulin FcγI and FcγII receptors (Lindenbach and Rice, 2001). An

alternative ADE mechanism that requires complement has also been described (Cardosa

et al, 1983; 1986).

The most common route of entry of flaviviruses into the cell is thought to be via

receptor-mediated endocytosis following studies with WNV, YFV, and KUNV in cell

cultures (Gollins and Poterfield 1985; Ishak et al, 1988; Ng and Lau, 1988). In this

process, following attachment of the virion on the host cell surface, invagination of the

cell membrane occurs which results in formation of coated vesicles that internalise the

virion and transport it to the cell cytoplasm (Gollins and Porterfield, 1985). Acid

CHAPTER 1

LITERATURE REVIEW

9

catalysed membrane fusion then takes place when E protein is exposed to the local mild

acidic conditions in the endosomes. This results in permanent structural changes of this

protein (reviewed in Heinz, 2003), causing uncoating of the membrane, release of the

nucleocapsid to the cytoplasm and eventually leading to the translation of the viral

genome. Alternatively, entry of flaviviruses by direct fusion at the plasma membrane

has also been observed following infection with JEV and DENV-2 in mosquito cells

and human monocytes (reviewed in Chambers et al, 1990a).

Translation occurs in association with the membrane of rough endoplasmic reticulum

(Lindenbach and Rice, 2001). Translation initiation is cap-dependent, starts at the 5’ end

of the viral genome and concludes at the stop codon located near the 3’ end to produce a

single polyprotein (Calisher and Gould, 2003; Alvarez et al, 2005). The polyprotein is

cleaved into 10 viral proteins. Host signal peptidase is associated with cleaving the

capsid-premembrane (C-preM), premembrane-envelope (prM-E), envelope-NS1 (E-

NS1) and near the C terminus of NS4A proteins. In contrast, the viral serine protease is

responsible for cleaving the NS2A/NS2B, NS2B/NS3, NS3/NS4A, NS4A/NS4B and

NS4B/NS5 (Lindenbach and Rice, 2003). Some of these proteins translocate to the

lumen of endoplasmic reticulum while the others remain localised on the cytoplasmic

site, depending on their roles in virus life cycle.

1.1.7 REPLICATION

Viral RNA replication occurs in the perinuclear region of the cells following translation

of viral RNA polymerase (Westaway et al, 1997). Although vesicle packets are the most

likely sites of viral replication, these structures are only observed late in infection and

thus it is still unclear where the initial RNA replication takes place (Lindenbach and

Rice, 2003). Replication starts with the production of negative strands, which serve as

templates for synthesising new genomes, positive strand RNA (Westaway et al, 1985).

Replication of flaviviruses has been shown to employ a semiconservative mechanism

with three major types of RNA detected. These include RNase sensitive 40-44S single

stranded virion RNA (vRNA), RNase resistant 20S double-stranded replicative form

(dsRF), and RNase partially sensitive 20-28S replicative intermediate form (RI)

(Lindenbach and Rice, 2001). All 3 species of viral RNA have been detected both in

flavivirus infected cells and mouse brains (Chu and Westaway, 1985; Urosevic et al,

1997a). DsRF serves as the template for viral RNA synthesis, in which the newly

CHAPTER 1

LITERATURE REVIEW

10

synthesised RNA strand replaces the existing positive strand RNA in the dsRF (Chu and

Westaway, 1985). The cycle period taken for a nascent RNA to displace the pre-existing

RNA in the RI is about 15 minutes (Westaway et al, 2002). The synthesis of viral RNA

is also shown to be asymmetric in vivo, with the production ratio of positive strand

RNA to the negative complementary strand is 10:1 to 100:1 (Brinton, 2001).

At present, regulation of RNA synthesis and the components of flavivirus replication

complexes have not been fully characterised (Lindenbach and Rice, 2001). However,

the involvement of both 3’ and 5’ UTR in the flavivirus genome replication has been

previously documented (Khromykh et al, 2001; Lo et al, 2003; Alvarez et al, 2005).

Deletion of RNA elements in the 3’ UTR has been shown to affect synthesis of DENV

RNA (Alvarez et al, 2005). The interaction of certain cellular proteins as well as viral

proteins such as NS3 and NS5 with the 3’terminal structures may be important in the

regulation of transcription (Brinton, 2001). A host cellular factor identified as

translation elongation factor-1 has been shown to interact with the 3’ stem loop

structure of the viral RNA during WNV infection (Blackwell and Brinton, 1997). In

vitro studies have also shown that interaction between cyclisation sequences (CS)

present in 3’ UTR and 5’ region of the ORF is required for RNA replication (You et al.,

2001).

1.1.8 VIRUS ASSEMBLY AND RELEASE

Upon completion of viral replication, virion morphogenesis takes place in association

with intracellular membranes. In flavivirus-infected cells, two modes of maturation have

been demonstrated; the trans-mode and the cis-mode (Hase et al, 1987). Maturation of

most flaviviruses is via the trans-mode and occurs in the lumen of ER (reviewed in

Lindenbach and Rice, 2001). In the cytoplasm, nucleocapsids or viral cores are formed

when the C protein interacts with genomic RNA. These structures then become

enveloped following budding process into the ER lumen (Lorenz et al, 2003; Lorenz

et al, 2002; Lobigs and Lee, 2004). Intracellular immature virions, which contain

heterodimers of E and prM proteins then accumulate within the membrane-bound

vesicles and are transported through the host secretory pathway (Mackenzie and

Westaway, 2001). Prior to release of mature virions, the glycans on E and prM are

altered followed by cleavage of prM in the trans-Golgi network by furin enzyme.

Virions are then transported to the cell surface membrane by secretory vesicles, where

CHAPTER 1

LITERATURE REVIEW

11

fusion occurs resulting in the release of progeny virions by exocytosis (reviewed in

Brinton, 2002; Lindenbach and Rice, 2001).

During cis-mode maturation, structural proteins are transported to the plasma membrane

for insertion at the budding site (Ng et al, 2001; Chu and Ng, 2002). Maturation occurs

at the plasma membrane and virions egress by budding rather than by exocytosis

process (Ng et al, 1994; reviewed in Ng and Chu, 2002). However, this mechanism has

been reported only in specific virus-host cell combinations including infection of

DENV-2 strain PR-159 in C6/36 cells (Hase et al, 1987) and WNV strain Sarafend in

Vero and C6/36 cells (Sreenivasan et al, 1993; Ng et al, 1994). Host microtubules and

actin filaments as well as the 5’ end of the viral genome have been shown to have a

critical role in the maturation and release of WNV Sarafend by this process (Ng et al,

2001; Chu et al, 2003; Li et al, 2005).

1.2 PATHOGENESIS OF FLAVIVIRUSES

Infection with flaviviruses in humans induces diseases ranging from non-specific febrile

illnesses (fever, headache, myalgia and malaise) to severe diseases such as encephalitis

and dengue hemorrhagic fever (DHF) (Tsai 2000; Mackenzie et al, 2002a). In Asia,

30,000 to 50,000 of JE cases with 10,000 deaths are reported annually (Burke and

Monath, 2001; Solomon et al, 2000). With WNV infection, diseases are mostly

asymptomatic with about 20-30% infected individuals suffering from symptomatic

diseases characterised by fever, headache, back pain and other minor symptoms.

(Mackenzie et al, 2004). KUNV-induced diseases however are rather mild and are

rarely associated with encephalitis (Hall et al, 2002). On the contrary, it has been

reported that during MVE infection, about 20% of clinical cases are fatal and 40% of

survivors suffer from permanent neurological sequelae (Mackenzie et al, 2002b).

In patients suffering from encephalitis, movement disorders such as tremor, and

Parkinsonian syndrome including rigidity and postural instability are commonly

observed. Some patients that survived flavivirus infections have been shown to suffer

from long-term cognitive and neurologic impairments (Mackenzie et al, 2004).

CHAPTER 1

LITERATURE REVIEW

12

Pathogenesis of flaviviruses in humans has not been fully characterised. At present,

vaccine or effective treatment is limited, thus further understanding of flavivirus-

induced illness is important to help design better therapy to treat the diseases. Infection

of flavivirus in rodents induces encephalitis; therefore this animal provides an excellent

model to study development of flavivirus-induced-encephalitis in humans. However,

certain diseases like DHF that is also inflicted on human do not cause similar disease in

mice; thus DHF phenomenon cannot be studied in these animals.

1.2.1 INFECTION IN VERTEBRATE HOSTS

During natural infection, flaviviruses are introduced into their vertebrate hosts by

mosquito or tick bites. The virus is then believed to replicate although not extensively in

the skin and infects Langerhan’s dendritic cells (reviewed in Chambers and Diamond,

2003). Infected dendritic cells then transport and spread the virus to the local lymph

nodes (Wu et al, 2000). The virus exits the lymph nodes via the efferent lymphatics and

later spreads to other organs such as liver, heart and kidney via the blood, establishing

systemic infection (Solomon and Vaughn, 2002). Following development of a sufficient

level of viraemia, virus can enter the brain via several routes. In weanling mice, the

olfactory mucosa has been shown to be the route for virus spread to the brain (Solomon

and Vaughn, 2002). WNV however was shown to spread to the central nervous system

(CNS) via the haematogenous route following footpad infection in mice (Diamond et al,

2003). Studies on the JEV and WNV-induced encephalitis in humans suggested that

hematogenous rather than olfactory route was a more probable way for the virus to get

disseminated to the brain (Desai et al, 1995; Shieh et al, 2000).

Means on how the virus crosses the blood brain barrier has also been studied. This

process could either occur through passive transport across the endothelium, replication

of virus in the endothelial cells or via infected inflammatory cells that act like ‘Trojan

Horses’ and carry the virus to the brain parenchyma (reviewed in Solomon and Vaughn,

2002). The virus species, vertebrate host and route of virus infection may influence

pathogenesis and virus spread to the brain, and this eventually will determine the

development or severity of encephalitis in infected host. During WNV infection, virus

replication in endothelial cells seems to be important while in contrast, JEV seems to

undergo endocytosis and transportation across endothelial cells and pericytes without

replicating in these cells, as observed in the brains of suckling mice (Liou and Hsu,

CHAPTER 1

LITERATURE REVIEW

13

1998). Axonal transport of flaviviruses to the olfactory lobe in the brain has been

suggested as an additional alternative pathway of flavivirus entry into the CNS. This

route is believed to be an important route of virus transport during i.n. infection of

WNV (reviewed in Chambers and Diamond, 2003). Peripheral inoculation of SLEV and

MVEV strains also result in the virus spread from the olfactory bulb, in a roastal to

caudal direction (reviewed in King et al, 2007).

In the CNS, neurons are the primary sites of viral replication (Hase et al, 1987; Silvia,

2004). In vitro infection and antigen production in astrocytes and oligodendrocytes has

been demonstrated but there is very little evidence that these glial cells are permissive to

virus infection in vivo (Jordan et al, 2000; Chen et al, 2000; Chambers and Diamond,

2003).

1.2.2 UP-REGULATION OF HOST CELL SURFACE MOLECULES UPON

FLAVIVIRUS INFECTION

Major histocompatibility complex (MHC) class I and class II molecules have important

involvement in shaping the adaptive immune response as they engage in antigen

presentation to the T cells. MHC class I and II molecules interact with and cause

activation of cytolytic T lymphocytes (mainly CD8+ T cells) and CD4+ helper T cells,

respectively (Abraham and Manjunath, 2006). Mouse MHC class I has a classical

region known as class Ia (H-2b, H-2K and H-2L) and a non-classical region identified

as class Ib (Shawar et al, 1994). Meanwhile, mouse MHC class II region consists of two

loci, I-A and I-E. In order to escape surveillance from CTL, some viruses induce down-

regulation of MHC I and II cell surface molecules (Diamond, 2003). However, reduced

expression of MHC I may result in infected cells being recognised and eventually killed

by natural killer (NK) cells. In contrast, infection by flaviviruses causes increased

expression of MHC I molecules on infected cells, as shown in various primary cells

types isolated from rats, mice, and humans upon infection with WNV, MVEV, JEV and

DENV (reviewed in King et al, 2003; Abraham and Manjunath, 2006). Up-regulation of

MHC I molecules by WNV was demonstrated to be due to an increase in MHC gene

transcription as well as an increase in peptide import into the lumen of the ER (Cheng et

al, 2004; Kesson et al, 2002; Mullbacher and Lobigs 1995; Momburg et al, 2001). Up-

regulation of MHC I and II expression is functional, causing infected cells to be more

susceptible to flavivirus-specific CTL and MHC I specific CTL (Kesson and King,

CHAPTER 1

LITERATURE REVIEW

14

2001) and triggering cytokine productions from MHC II- specific effector T cells (Liu

et al, 1989b). However, expression of MHC I is dependent on the cell cycle as well as

adhesion status, with cells infected at G0 phase showing higher up-regulation of these

cell surface molecules than cells infected in other phases of the cell cycle (Shen et al,

1995a; 1995b). In addition to MHC molecules expression, adhesion molecules

including ICAM-1, VCAM-1 and E-selectin were also shown to be increased following

flavivirus infections (King et al, 2003).

It has been suggested that induced up-regulation of MHC I molecules on infected cells

is a mechanism used by flavivirus to evade NK cells surveillance early in infection. This

perhaps would give adequate time for the virus to replicate at high titres and induce

prolonged viraemia and subsequently would allow invasion of the CNS (Momburg et al,

2001; Diamond et al, 2003a). Additionally, greater expression of MHC I also increases

the avidity of interaction between infected cells and virus-specific T cells (Kesson et al,

2003). Given that cells in G0 phase have higher induction of MHC I, these cells appear

to be more susceptible to killing by low affinity T cells. This in turn, will allow infected

cells in other phase cycle to escape T cells detection and maintain a low immunological

profile but still manage to spread the infection (Kesson et al, 2002; King et al, 2003).

However, it is also possible that the flavivirus-induced MHC I expression is a by-

product of virus replication and not a strategy to evade the immune system (Lobigs,

2003b).

1.2.3 CENTRAL NERVOUS SYSTEM DISEASES

1.2.3.1 Nature and properties of the central nervous system

Until recently, the brain was thought to be an immune privileged organ and that there is

no communication between the brain and the immune system except during infection or

injury. This assumption is due to several observations: the presence of a tight layer of

endothelial cells known as blood brain barrier (BBB) in the brain which prevents free

movement of cells and proteins from the periphery into the brain; the lack of antigen

presenting cells (APC) and expression of MHC cell surface molecules in the CNS; the

lack of a classical lymphatic drainage system; prolonged survival of tissue grafts within

the CNS; and the presence of neurons that are post-mitotic and non renewable cells

(Bradl and Frugel, 2002; Reiss et al, 2002). However, now it has become evident that

CHAPTER 1

LITERATURE REVIEW

15

the CNS is in fact under constant immune surveillance both during healthy and infection

stages and thus can react accordingly upon any insults or injury. Activated T cells can

enter the CNS but are not retained in the absence of antigen; they either leave the CNS

or die in situ (Binder and Griffin, 2003).

The CNS consists of neurons and glial cells including microglia/macrophages,

astrocytes and oligodendrocytes. The neurons’ basic function is to receive and transmit

electrochemical signals via synaptic connections (Redwine and Evans, 2002). They are

long-lived, terminally differentiated and non-replaceable cells (Griffin, 2003). Microglia

are macrophage-like cells that comprise up to 20% of total glial cells (Gehrmann, 1996).

Immune surveillance is one of the primary functions of microglia and thus they are the

first cell type to respond to CNS injuries or insults (Gehrmann, 1996; Redwine and

Evans, 2002). Invading pathogens are rapidly recognised by microglia, which in turn

produce pro-inflammatory cytokines (such as IL-1, TNFα and IL-6) and eventually

initiate an inflammatory cascade (Haschish, 2002). Unlike other resident brain cells,

microglia are derived from the bone marrow precursors, which invade the CNS at an

early stage of embryonic development to give rise to brain macrophage-like cells

(Becher et al, 2000). Oligodendrocytes form and maintain the myelin that surrounds the

axons of neurons and infection of these cells usually leads to demyelinating diseases.

Astrocytes are the supporting cells for neurons that secrete neuroprotective factors and

neurotransmitters, and are responsible for removing toxic materials (Griffin, 2003). All

brain resident cells are capable of producing cytokines and chemokines (Redwine and

Evans, 2002).

It is still not clear which cells take up the role of the APCs in the brain parenchyma.

Among cells considered to be possible APCs are microglia/macrophages, astrocytes and

oligodendrocytes. However, to act as an APC, particular cells must be able to express

MHC II cells surface molecules, integrins, CD40, co-stimulatory molecules B7 and

cytokines such as IL-12 (Reiss et al, 2002). The strongest data indicated that microglia

possess these characteristics, thus suggesting the predominant role of these cells as APC

in the CNS (Reiss et al, 2002). Expression level of B7 and B7-2 has been shown to be

significantly higher in activated microglia than in astrocytes (Satoh et al, 1995; Soos et

al, 1999).

CHAPTER 1

LITERATURE REVIEW

16

In normal, uninfected brains, there is a lack of immunological activity. Microglia and

other cells are usually kept in a quiescent state through production of neurotrophins and

anti-inflammatory TGFβ (Griffin, 2003). Expression of MHC molecules is also either

minimal or absent and confined to only dendritic cells and macrophages at the

meninges, choroid plexus and perivascular spaces (Binder and Griffin, 2003; Schneider-

Schaulies et al, 2000). MHC II is expressed minimally in microglia and possibly

astrocytes (although this is less evident) but can be up-regulated upon infection as well

as by the trafficking of activated T cells through the CNS. Generally, neurons are

thought not to express MHC I cell surface molecules as these cells are non-replaceable

and thus, lysis of neurons by cytolytic T cells will lead to detrimental and fatal

outcomes of disease (Binder and Griffin, 2003).

When infectious agents reach the CNS, disease only develops if a sufficient number of

cells is infected, resulting in brain dysfunction. CNS tissue is unique in that it has a high

metabolic rate and a low regenerative capacity. Some viruses specifically target neurons

as their site of replication. However, not all types of neurons are equally susceptible as

this depends on factors such as tissue tropism, route of virus entry as well as virus

virulence (Griffin, 2003). Neuronal tissue can be damaged or completely destroyed by

intracellular replication of viruses. Poliovirus, a positive single stranded RNA virus that

causes acute infection, replicates in the anterior and posterior horn cells of the spinal

cord resulting in flaccid paralysis. In contrast, Rabies virus causes non-lytic infection of

the neurons but disease develops because the virus interferes with neuronal cell

functions in vital centres that regulate sleep, body temperature and respiration

(Schneider-Schaulies et al, 2000). The glial cells; oligodendrocytes, astrocytes and

microglia can also be infected, depending on the type of infecting viruses.

In general, clearance of virus from the brain depends on the type of cells being infected

in the brain as well as the type of infecting virus. Viral elimination from infected

neurons usually involves a non-cytolytic mechanism as neurons are non-renewable

cells, resistant to apoptosis and do not express MHC I molecules. In contrast, efficient

clearance of virus from glial cells may involve NK or T cell-mediated cytolytic

mechanisms (Binder and Griffin, 2003).

CHAPTER 1

LITERATURE REVIEW

17

1.2.3.2 Flavivirus infections in central nervous system

In the brain, neurons are the principal target for flavivirus infection as has been

documented in vivo with several flavivirus infections in mice (Hase et al, 1990;

Andrews et al, 1999; Silvia et al, 2004). Localisation of viral antigen is also evident

around neurons but not in other CNS resident cells during fatal human cases of JEV

infection (Johnson et al, 1985). Infection with JEV and MVEV in mice caused

cytoplasmic rarefaction in the neurons (Hase et al, 1990b; Silvia 1999). The cytoplasm

appeared to be round and empty with a nucleus in the centre. By electron microscopy,

virus was shown to be replicating in the cell secretory pathway including RER and

Golgi apparatus of the neurons, eventually causing cytoplasmic organelle damage and

rarefaction as seen by light microscopy (Hase et al, 1990a). Infection of other cell types

in the brain is less clear. Recently, human primary cell cultures of neurons, astrocytes

and microglia were infected with WNV and virus growth was observed only in neurons

and astrocytes. Microglia however failed to support WNV replication although these

cells did produce cytokines upon infection (Cheeran et al, 2005). No infiltrating

leucocytes, endothelium or perivascular cells were shown to be infected during JEV or

MVEV infection (Hase et al, 1990b; Andrews et al, 1999).

Neuronal infection by viruses typically induces host inflammatory responses. Typical

inflammatory responses seen following flavivirus infection in the CNS include

perivascular cuffing and cellular infiltrates accumulation at the meninges and brain

parenchyma as well as microglia nodules formation associated with neuronophagia

(King et al, 2003; Chambers and Diamond, 2003). Among the leucocytes that present

in the brain are neutrophils, macrophages and lymphocytes although the composition,

type and importance of these infiltrating cells depend on the experimental models used

(Andrews et al, 1999; Silvia et al, 2004). Migration and infiltration of perivascular

leucocytes is the result of production of IFN type I from infected neurons, which then

signals microglia and astrocytes to produce secondary cytokines and chemokines to

attract these inflammatory cells to the brain (Gouwy et al, 2005).

Apoptosis is an active process of cell death that has an important role in development,

morphogenesis, tissue remodelling and immune regulation. This process also has been

associated with the pathogenesis of many diseases (Hay and Kannoroukis, 2002). Cells

undergoing apoptosis have distinct morphological features including cytoplasm

CHAPTER 1

LITERATURE REVIEW

18

condensation, fragmentation of the cell nucleus and chromatin condensation, and

membrane blebbing (Courageot et al, 2003). The biochemical hallmarks of apoptosis

include activation of endonucleases, DNA degradation into oligonucleosomal fragments

and activation of caspases (Courageot et al, 2003). In vivo and in vitro studies have

demonstrated that certain flaviviruses are capable of inducing apoptosis. The molecular

mechanism for the occurrence of apoptosis is not fully defined but it has been suggested

that some flavivirus proteins such as the capsid and NS3 may be directly involved in

inducing neuronal cell death (Chamber and Diamond, 2003; Prikhod’ko et al, 2002).

St. Louis encephalitis virus (SLEV) infection in human mononuclear cells, K562 and

mouse neuroblastoma cell line, Neuro2a resulted in up-regulation of pro-apoptotic bax

gene leading to apoptosis (Parquet et al, 2002). In addition, apoptosis also has been

reported during in vitro infection by Langat virus, JEV, WNV and DENV (Prikhod’ko

et al, 2002; Liao et al, 1997; Chu and Ng, 2002; Despres et al, 1996). In newborn mice,

DENV was used to study induction of apoptosis. Infected mouse brain tissue was

assayed for both viral antigens and apoptosis, and neurons were shown to be both

positive for DENV infection as well as apoptosis (Despres et al, 1998). In contrast,

infection with viruses such as JEV and MVEV in rodents did not cause any apoptosis

Hase et al, 1990; Silvia 1999; Andrews et al, 1999, and a fatal outcome of infection was

attributed possibly to neuronal dysfunction (Hase et al, 1990).

1.2.4 NEUROINVASIVENESS AND NEUROVIRULENCE

Infection of flaviviruses in different individuals may result in different disease

outcomes. This is because the severity of the diseases induced by these viruses relies on

many underlying factors and one of them is the type of infecting virus. While some

flaviviruses can induce fatal encephalitis, others only cause asymptomatic infections in

the same host, an outcome that is influenced by the neuroinvasiveness and

neurovirulence properties of the viruses.

Neuroinvasiveness is the ability of the virus to enter and infect cells or tissue in the CNS

upon peripheral inoculation. Neurovirulence on the other hand is the capacity of the

virus to infect and replicate in the CNS (Schneider-Schaulies et al, 2000). Early studies

on molecular determinants of virulence in flaviviruses were done by comparing the

genomic sequences of high and low virulence strains of YFV, JEV, DENV and tick

borne encephalitis virus (TBEV) (reviewed in Hurrelbrink and McMinn, 2003).

CHAPTER 1

LITERATURE REVIEW

19

However, more specific information on neuroinvasiveness and neurovirulence of

flaviviruses has been obtained from studies using various approaches such as single

plaque purification of uncloned viruses, selection of neutralisation escape variants,

limited passage of the virus in cell culture or mice and site-directed mutagenesis of viral

infectious clones (reviewed in Hurrelbrink and McMinn, 2003). In animal studies,

neuroinvasiveness is tested by inoculating the virus via the peripheral route while

neurovirulence is examined by inoculating the virus directly into the brain. Some

flaviviruses are both neuroinvasive and neurovirulent while others may be neurovirulent

but not neuroinvasive.

Studies with viruses such as MVEV, TBEV and JEV using variants selected either from

neutralisation-escape mutants or by serial passage in cell cultures have demonstrated

that the E protein plays an important role in the neuroinvasiveness of flaviviruses

(reviewed in McMinn, 1997; Lobigs et al, 1990). Alteration of amino acid residues in E

protein may not only change the receptor binding but may also result in reduced virus

uptake into the cells following initial attachment and change in pH-depended fusion

activity, all of which may cause the loss of neuroinvasiveness of certain viruses

(McMinn, 1997). McMinn and co-workers (1996) have shown that MVEV variants with

low neuroinvasiveness have an altered amino acid at residue 227 in E glycoprotein.

An in vivo study with the New York strain of WNV revealed the association of

glycosylation of E protein with neuroinvasiveness of the virus. Upon subcutaneous (s.c.)

infection with glycosylated and non-glycosylated variants of WNV, the former variant

caused higher mortality in mice although mortality following i.c. infection did not differ

between these two variants (Shirato et al, 2004a). On the contrary, WN25 and WN25A,

that were isolated from Israel, had glycosylated E proteins but were not neuroinvasive

when inoculated intraperitoneally (i.p.) into mice (Halevy et al, 1994). Thus, although

evidence from both in vitro and in vivo studies implicated the role of E protein in

neuroinvasiveness of the same virus strains, this notion cannot be generalised for all

flaviviruses. Studies with birds revealed that neuroinvasive viruses such as WNV strain

NY99 were capable of eliciting high levels of viraemia and eventually death of the

infected host. This is in contrast to infection with KUNV, which is non-neuroinvasive

and induces low viraemia levels and minimal morbidity in infected birds (Brault et al,

2004).

CHAPTER 1

LITERATURE REVIEW

20

In addition to the E proteins, the loss or reduced neuroinvasiveness of flaviviruses has

also been associated with the NS1 protein mutations. An amino acid mutation, from

proline to leucine at residue 250 in the NS1 of MVEV and KUNV was demonstrated to

cause attenuation of these viruses in weanling mice during i.p. challenge (Clark et al,

2007; Hall et al, 1999).

The blood brain barrier (BBB) is a tight barrier formed by endothelial cells and acts as a

physical barrier that restricts movement of large molecules (Abbott et al, 2006). The

breakdown of the BBB however can facilitate the entry of non-neuroinvasive virus to

the brain. The permeability of this barrier can be altered by cytokines such as IL-1, IL-8,

TNF and IFN as well as cytolytic T cells (Charturvedi et al, 1991; Mathur et al, 1992;

Licon Luna et al, 2002). In addition, physical stress and the use of chemicals such as

dimethyl sulphoxide (DMSO), sodium dodecylsulphate (SDS), lipopolysaccharides

(LPS) and polyinosinic:polycytidylic (pIc) can also contribute to the breakdown of BBB

(Kobiler et al, 1989; Abbott et al, 2006; Haahr, 1971).

Neurovirulence of flaviviruses is probably determined by structural proteins such as E

protein, as well as non-structural proteins. In DENV-4, neurovirulence determinants are

located in the prM/E gene region (Pletnev et al, 1992). Other studies have shown that

NS1 mutation led to reduced virus replication and loss of neurovirulence of DENV and

YFV (reviewed in McMinn, 1997). In MVEV variants selected by serial passage in

Vero cells, loss of virulence was due to mutation in the non structural region and/or

3’ UTR (McMinn et al, 1995).

1.3 IMMUNE RESPONSE AND IMMUNOPATHOLOGY

Invasion of pathogens will provoke the immune system as part of the host defence

mechanism. There are two parts of the immune system that are induced and may

interact with each other during infection. These are innate and adaptive immunity.

Innate immunity, which has limited capacity to distinguish different pathogens, mounts

a rapid non-specific antimicrobial response within hours of infection (Nash and

Usherwood, 2000). In some cases, the action of innate immunity alone may be adequate

to abolish an infection. However, if this early host defence in preventing infection is

unsuccessful, it can still slow down the spread of infection before another branch of the

CHAPTER 1

LITERATURE REVIEW

21

immune system, the adaptive immunity becomes active. The adaptive immune response

is a much slower response but with high specificity for the microbes (Nash and

Usherwood, 2000). The high specificity for different infectious agents allows this

branch of the immune system to use several discrete mechanisms to combat different

types of infections (Nash and Usherwood, 2000).

A vigorous immune system however may act as a double-edged sword to the host. In

order to provide protection from the harmful effect of infectious agents, it may at the

same time induce collateral damage to host cells as well (Lucas et al, 2006). Hence, an

infected host may not die directly from virus infection, but from immunopathological

diseases caused by one or several components of the immune system (Santana and

Rosenstein, 2003).

Many studies have been undertaken to look at the role of both the innate immune

system and adaptive immunity in protection as well as in pathogenesis of flavivirus

infection.

1.3.1 INNATE IMMUNE SYSTEM

1.3.1.1 Macrophages

Macrophages are mononuclear phagocytes derived from blood monocytes and are

involved in non-specific host defence. Blood monocytes migrate to various tissues and

organs and differentiate to macrophages in response to specific stimuli (Beutler, 2004).

Macrophages can also act as APCs and capable of producing cytokines such as IFN

and TNF, which can contribute to the immunoprotection of the infected host. Another

protective and antiviral effect provided by macrophages is the induction of nitric oxide

synthase-2 (NOS-2) following IFN activation which results in the production of NO

and other reactive oxygen intermediates (Saxena et al, 2000). Macrophages can also

clear virus using several mechanisms including phagocytosis or by opsonisation of

virus-antibody complexes via the Fc receptor.

Selective depletion of macrophages with silica was shown to increase the susceptibility

of mice to YFV-induced diseases (Zisman et al, 1971). Silica impairs liver macrophages

function thus resulting in slower viral clearance and prolonged viraemia. Virus can then

CHAPTER 1

LITERATURE REVIEW

22

enter the brain earlier and undergo rapid replication before an antibody response is

developed. The important role of macrophages in host defence has also been

demonstrated following challenge of a non-neuroinvasive variant of WNV in mice

(WN-25) (Ben-Nathan et al, 1996). This attenuated WNV did not cause any mortality in

control mice. However, following depletion of macrophages using the drug

dichloromethylene diphosphonate (clodronate) in the mice, 70-75% mortality was

observed when mice were challenged with WN-25 (Ben-Nathan et al, 1996).

Although macrophages are an important host innate immunity agent, they can also have

immunopathological role during flavivirus infections. In vivo and in vitro studies have

shown that macrophages can be infected and can contribute to virus pathogenesis by

antibody dependent enhancement (ADE) (Cardosa et al, 1986; Pantelic, 2005;

Anderson, 2003). In vitro macrophage infection has been shown to depend upon

several factors including mouse strain, type of infecting virus and different macrophage

populations (Cardosa et al, 1986; Kreil et al, 1997). Thioglycollate is a rich nutrient

medium that has been used widely to induce sterile peritoneal inflammation in order to

elicit macrophages. Intraperitoneal injection of thioglycollate increases peritoneal yield

10-fold with 80% of total cell population consisting of macrophages (Silvia et al, 2001).

Thiogycollate-elicited-macrophage cultures have been used in this and other laboratory

as an in vitro model of infection to study flavivirus pathogenesis.

1.3.1.2 Nitric oxide

Nitric oxide is a free radical gas, which has antimicrobial activity, and is derived as a

by-product during the conversion of L-arginine to L-citrulline by the NO synthase

enzyme (Bogdan, 2001; Licinio et al, 1999). There are three forms of this gene: NOS-1

is expressed in the neurons, NOS-2 is found in macrophages and NOS-3 is expressed in

endothelial cells (Nathan, 1992). NOS-1 and NOS-3 were mapped to distal and

proximal regions of murine chromosome 5, respectively (Gregg et al, 1995; Lee et al,

1995). The NOS-1 and NOS-3 are constitutively expressed while NOS-2 is inducible

(Reiss and Komatsu, 1998). IFN has been shown to be a potent inducer of NOS-2. In

many virus infections such as Sindbis virus and Herpes Simplex virus type I, NO has an

important role in inhibiting viral replication, promoting viral clearance and eventually

host recovery (Reiss and Komatsu, 1998). In flavivirus infection, NO activity

contributes both to recovery and immunopathology, depending on the type of infecting

CHAPTER 1

LITERATURE REVIEW

23

virus. During in vitro infection with JEV, NO caused inhibition of viral RNA, viral

protein accumulation and virus release from infected cells. In mice infected with the

same virus, inhibition of NO resulted in increased mortality, demonstrating the antiviral

role played by NO in JEV infection (Lin et al, 1997). The pathogenesis of TBEV in

contrast was shown to be partially mediated by NO. Blocking of its production by

administration of a competitive inhibitor of NOS-2 increased the mean survival time of

infected mice although they eventually succumbed to the infection (Kreil and Eibl,

1996). It was suggested that NO production increased cerebral blood flow and

peripheral inflammatory cell recruitment, which eventually led to the BBB breakdown.

1.3.1.3 Natural killer cells

Natural killer (NK) cells have the ability to kill infected cells by releasing granzymes

and perforin or by binding to the death receptors on the cell surface. A decrease in MHC

class I expression on target cells prompts NK cell activation by attenuating the

inhibitory signals. NK cell dependent lysis and antibody dependent mediated

cytotoxicity against DENV-infected cells has been observed (Kurane et al, 1984).

However, the bulk of evidence suggests that flaviviruses evade the NK surveillance by

up-regulating the expression of MHC class I molecules as well as adhesion molecules

(King and Kesson, 1988). Consistent with this, splenocytes from WNV-immunised

mice had poor NK cell lytic activity (Momburg et al, 2001) and mice that were

genetically deficient in NK cells did not exhibit increased morbidity or mortality rate

compared to wild type control mice during flavivirus (Chambers and Diamond, 2003).

1.3.1.4 Neutrophils

Neutrophils are also known as polymorphonuclear cells and respond rapidly to

chemotactic stimuli and are activated by cytokines (Nash and Usherwood, 2000).

Mortality following peripheral infection of MVEV strain BH3479 in weanling Swiss

mice is associated with development of encephalitis and inflammatory responses, which

is predominated by the presence of neutrophils (Andrews et al, 1999). The infiltration of

neutrophils was preceded by an increased mRNA expression of TNFα and the

neutrophil-attracting chemokines N51/KC which is a murine homolog to human IL-8.

The mRNA expression of inducible NOS (iNOS) coincided with the presence of

neutrophils in the brain and onset of encephalitis. Depletion of neutrophils with

CHAPTER 1

LITERATURE REVIEW

24

monoclonal antibodies resulted in higher survival although similar viral titres in the

brains were detected as in the brains of control mice. Neutrophils possibly induce their

deleterious effect in the CNS by producing NO and other inflammatory mediators,

which may disturb CNS homeostasis and neuronal functions (Andrews et al, 1999).

1.3.2 ADAPTIVE IMMUNITY

There are two branches of adaptive immunity based upon the major components that

mediate the response; humoral and cell mediated immunity. Humoral immunity mainly

involves antibodies (produced by B cells) which recognise as well as eliminate antigen

while cell mediated immunity involves T lymphocytes (Becher et al, 2000). In an

infected host, the bias to mount either one of these types of specific immune response is

dependent on CD4+ helper T cells. In order for CD4+ cells to mediate the effect, they

must recognise the antigenic peptide/MHC II complexes and co-stimulatory molecules

present on the cell surface of APCs (reviewed in Mullbacher et al, 2003). Following

this, CD4+ helper T cells then become activated and differentiate into two major

subpopulations that differ in the cytokine profiles that they secrete (Becher et al, 2000).

T helper 1 (Th1) cells secrete IL-2, IFNγ and TNFβ and thus mediate a cellular immune

response through activation of macrophages and cytotoxic T cells. Th2 cells on the

other hand secrete IL-4, IL-5, IL-6, IL-10 and IL-13, leading to the maturation of B

cells and degranulation of mastocytes, therefore inducing the humoral immune response

(Santana and Rosenstein, 2003).

The role of both humoral and cell mediated immune responses in either protection or

immunopathology of flavivirus infections have been investigated mostly using murine

model utilising different viruses and routes of inoculation. B and T cells have been

shown to have different roles during disease development in different experimental

models.

CHAPTER 1

LITERATURE REVIEW

25

1.3.2.1 Humoral mediated immunity

Many studies have been undertaken to study the role of humoral immune response in

protecting infected host from fatal flavivirus infection. Antibody mediates its effect via

virus neutralisation, which inhibits virus attachment, prevents release of cell-bound

virus or blocks the fusion step associated with un-coating of virus particles (Hooper

et al, 2002). All of these steps result in protecting cells from being infected. However, if

virus has already entered the cells, antibody can clear the virus through complement-

mediated cell lysis or antibody-dependent cytotoxicity that results in cell lysis (Hooper

et al, 2002). Antibody may also utilise a non-lysis mechanism to rid the virus from cells

such as by inhibiting viral RNA transcription and restoring antiviral function of IFN in

infected cells (Dietzschold et al, 1992).

Following flavivirus infection, neutralising antibodies (NA) are mostly induced by the

E protein that has antibody eliciting epitopes widely distributed over its surface (Heinz,

1986; Roehrig et al, 1989). Non-neutralising yet protective antibodies on the other hand

have been reported to recognise the NS1 proteins. Immunisation with recombinant or

purified NS1 has been shown to be protective against flavivirus challenge (Calvert et al,

2006; Hall et al, 1996). Following in vitro and in vivo studies, protection by anti-NS1

antibodies has been previously reported to operate via the Fc receptor dependent

mechanism (reviewed in Brinton et al, 1998). However, Chung and co-workers (2006)

have recently demonstrated that distinct regions of NS1 could also elicit humoral

immunity through an Fc receptor independent mechanism. Other viral proteins that are

also immunogenic for humoral mediated immunity are C, prM, NS3, NS4B and NS5

(Brinton, 1998). Induction of both neutralising antibodies and non-neutralising

antibodies have been reported following infection with several flaviviruses including

JEV, MVEV, DENV, and YFV (Kurane, 2002; Konishi et al, 1995; Chambers and

Diamond, 2003).

Antibodies usually can be detected 4-6 days following peripheral infection and usually

associated with the termination of viraemia and the presence of virus in the brain (Bhatt

and Jacoby, 1976; Diamond et al, 2003b). During JEV infection, hemagglutination

inhibition (HI) antibody and NA were first detected in 1 week and persisted up to

5 weeks (Mathur et al, 1983). IgM antibodies usually predominate early in infection

while IgG antibodies appear later (Monath and Borden, 1971). IgM and IgG antibodies

CHAPTER 1

LITERATURE REVIEW

26

are important in controlling flavivirus infection, with the former antibodies involved in

preventing viral dissemination early in infection and in induction of IgG antibodies

production. IgG antibodies are then involved in shaping up the adaptive immune

response (Diamond et al, 2003b; Engle and Diamond, 2003).

The important role of humoral immunity in flavivirus infection became evident when

mice were treated with cyclophosphomide; a chemical that suppresses B cells and to a

lesser extent T cells prior to virus infection. The immunosuppressed mice did not

develop detectable anti flavivirus antibodies and this resulted in extended viraemia,

higher CNS viral burden and increased mortality (Bhatt and Jacoby, 1976; Camenga

et al, 1974; Cole and Nathanson, 1968). Diamond and co-workers (2003b) have further

defined the importance and specific role of antibody using wild type and genetically

deficient mice. Upon infection of low passage WNV via the footpad, wild type mice

developed viraemia on day 2 p.i. but the virus became undetectable on day 6 p.i. In

contrast, B cell deficient (µMT) mice had prolonged viraemia that lasted up to day 8 p.i.

In addition, viral titres in peripheral organs and the CNS were also much higher in µMT

mice and all died from WNV-induced encephalitis. When these mice were given heat-

inactivated serum from infected and immune wild type mice, they were protected from

the disease although the protection was not long lasting. Protection was shown to

depend on the initial amount of the immune serum given but eventually all mice

succumbed to viral infection when the antibody level had waned, with some mortality

observed as late as 60 days after infection (Engle and Diamond, 2003). This was in

contrast to wild type animal in which passive administration of immune serum

completely protected them from flavivirus infection and no mortality was observed even

after several months. In µMT mice, virus probably still replicated at low level and

persisted following administration of immune serum. When antibody titres eventually

declined, virus could replicate at higher levels again and subsequently induce fatal

encephalitis in the animal (Engle and Diamond, 2003). This study demonstrated that

antibodies are important in preventing virus dissemination and viraemia but they are not

sufficient to completely clear the virus.

Following brain infection with other viruses, antibodies have been shown to be pivotal

in clearing Rabies virus, mouse hepatitis virus (MHV), Theiler’s murine

encephalomyelitis virus (TMEV) and Sindbis virus from neurons in the CNS, while

CD8+ T cells only played an auxiliary role in some of these virus infections (Binder and

CHAPTER 1

LITERATURE REVIEW

27

Griffin, 2003; Kimura and Griffin, 2000). In neuronal infection with Sindbis virus,

inhibition of viral replication occurs when antibodies bind to the viral glycoproteins on

the cell surface membrane (Nash and Usherwood, 2000). In some studies, IFN type I

has been reported to act synergistically with antibodies to inhibit viral replication

(Griffin, 2003).

Although humoral immunity has a protective role in many virus infections, it can also

have a detrimental effect and contribute to the pathogenesis in some virus-induced

diseases. DHF is a classical example of an immunopathological disease, which is

humoral immunity-mediated. There are four DENV serotypes and individuals who have

had a previous infection with one DENV serotype have a higher risk of developing DHF

following secondary infection with a different DENV serotype than individuals without

prior infection (reviewed in Mackenzie et al, 2004). This disease may be caused by

ADE, when subneutralising antibodies from one serotype of DENV enhance the

infectivity of another DENV serotype for macrophages and monocytes by facilitating

the uptake of virus-antibody complexes via the Fc receptor (Burke and Monath, 2001).

This may increase the number of infected cells and eventually increase the production

of virus. Broom and co-workers reported a similar ADE phenomenon in mice infected

with MVEV (2000). When 3 weeks old mice received immune sera from mice

previously infected with sublethal dose of MVEV and JEV prior to MVEV challenge,

these mice were protected from fatal outcomes. However, when similar experiments

were performed using immune sera obtained upon sublethal challenge with KUNV or

passively immunised with JEV vaccine, mice succumbed to MVE infection. In fact,

mice passively immunised with JEV vaccine died more rapidly than the control group,

which received non-immune serum. This suggests that ADE may be involved in

accelerating disease development, as it is known that immunisation with live virus

induces higher levels of antibody than inactivated vaccine (Broom et al, 2000).

1.3.2.2 Cell-mediated immunity

Cell-mediated immunity (CMI) is mediated by T lymphocytes, CD4+ and CD8+ T

cells. T lymphocytes are initially naive cells and they require priming for the induction

of their effector functions (Santana and Rosenstein, 2003). APCs process antigenic

peptides for presentation by MHC II molecules, which are then recognised by CD4+ T

cells. In contrast, CD8+ T cells recognise MHC I/peptide complexes that are presented

CHAPTER 1

LITERATURE REVIEW

28

on infected cells. Activation of T cells consequently results in induction of effector

activity of T cells; which involves cytolysis of infected cells and/or control of infection

via cytokine production (Mullbacher et al, 2003).

The CMI has been suggested to have a more important role than humoral immune

response in recovery from flavivirus infection (Bhatt and Jacoby, 1976). T cells induced

following flavivirus infection or immunization usually have broad cross-reactivity,

although this also depends on the genetic background (MHC haplotypes) of the

experimental animals used (Hill et al, 1992; Kulkarni et al, 1992; Kurane, 2002;

Chambers and Diamond, 2003). The differences in MHC haplotyes also influence the

multiple epitopes recognised by T cells. For example, the CTL from BALB (H-2d) mice

recognise E protein while C57BL/6 mice (H-2b) and C3H mice (H-2k) mainly

recognise the NS1 protein during JE infection (Takada et al, 2000). In addition to

genetic background of the mice, virus strain could also affect the antigenic peptide

recognised by T cells. During MVEV infection, NS3 protein was the dominant source

of antigenic peptide for cytotoxic T cell recognition (Lobigs et al, 1994)

Previous studies using mice with suppressed T cells functions clearly showed the

importance of CMI in clearing flavivirus and to some extent preventing mortality in

infected hosts (Camenga et al, 1974; Nathanson and Cole, 1974). Adoptive transfer of

immune spleen cells conferred protection to mice against flavivirus infections

(Camenga et al, 1974; Mathur et al, 1983; Murali-Krishna et al, 1996; Desai et al,

1997). In JEV infection, when mice were given immune spleen cells lacking either T or

B cells prior to intracerebral virus challenge. Protection from encephalitis was abrogated

by depletion of T cells but not B cells (Mathur et al, 1983).

Many recent studies were undertaken to define the role of individual cells of the CMI

especially CD8+ T cells in immunopathology or protection following flavivirus

infection. However the role of these cells is very much influenced by factors such as the

strain of mice, type of infecting virus as well as the route of virus inoculation. In mice

that were previously immunised and then challenged i.c. with neuroadapted YF strain

17D, B cell, CD4+ and CD8+ deficient mice had 0%, 6.6% and 85% survival rates

respectively, despite the higher levels of CD8+ T cells detected in the brains of wild

type mice relative to CD4+ T cells (Liu and Chambers, 2001). This indicated that

although CD8+ T cells are functional during YFV clearance, they are not critical for

CHAPTER 1

LITERATURE REVIEW

29

survival of these mice and that antibody response driven by CD4+ is probably the

mechanism involved in conferring protection from the fatal outcome (Liu and

Chambers, 2001). Studies with other viruses such as the JHM strain of neurotrophic

mouse hepatitis virus also revealed the importance of CD4+ T cells in virus clearance

(Stohlman et al, 1998). During i.c. infection of this virus, there were more CD8+ T cells

infiltrating the brain in comparison to CD4+ T cells. However, the expression of CD8+

CTL effector function in the brain parenchyma is CD4+ dependent, and the absence of

CD4+ T cells in the CNS resulted in rapid apoptosis of CTL cells. Murali-Krisha and

co-workers (1996) have demonstrated that both CD4+ and CD8+ T cells are required

for protection against JEV infection in mice. In contrast, studies with WNV isolated in

New York indicated that CD8+ T cells have a more important role in animal recovery

(Diamond et al, 2003a; Shrestha and Diamond, 2004). Mice that lack CD8+ T cells

were more prone to the disease and had increased mortality following subcutaneous

WNV challenge. In these mice, although brain viral production is much higher than

wild type mice, absence of CD8+ T cells did not have any effect on antibody response

as well as the kinetics or magnitude of viraemia. In some CD8+ T cell deficient mice

that survived the infection, infectious virus could be recovered from the brain several

weeks later, indicating the pivotal role played by this subset of T cells in clearing the

virus and preventing persistent infection (Shrestha and Diamond, 2004). During

infection with viruses other than flaviviruses, impaired CD8+ T cell response has been

reported to affect clearance of Lymphocytic choriomeningitis virus (LCMV) but not

Influenza, Sendai or Vaccinia viruses (Mangada et al, 2002).

Intravenous (i.v.) infection of WNV Sarafend in mice showed the dual role of CD8+ T

cells; in recovery and immunopathology during virus infection (Wang et al, 2003b).

When mice were infected with either low or high doses of virus, cellular infiltrates

detected were predominantly CD8+ T cells and not CD4+ T cells. While CD8+ T cells

contributed to the recovery in mice infected with the low WNV dose, this cell type

caused immunopathology in mice challenged with the high WNV dose. At high dosage,

i.v. infection resulted in 100% mortality of infected mice. In contrast, virus challenge in

CD8+ T cell deficient mice increased mean survival time and reduced mortality,

indicating that CD8+ T cells contributed to the pathogenesis of WNV. In addition,

several other studies have shown that cross-reactive T cells could lead to DHF by

causing tissue damage and production of cytokines that aggravate vascular leakage

(Loke et al, 2001). It has been suggested that following secondary DENV infection, the

CHAPTER 1

LITERATURE REVIEW

30

pre-existing low avidity memory T cell population expands at a higher rate than the

naive high avidity T cell population resulting in deregulation of immunological

response to DENV infection (Navarro-Sanchez et al, 2005). Children suffering from

DHF were found to have elevated levels of soluble CD8+ T cells in the serum in

comparison to children suffering from dengue fever (DF) only (Kurane et al, 1991).

Upon recognition with MHC I proteins complexed with short viral peptide, T cells

become activated and mediate their effect through two independent effector functions;

production of IFNγ or cytolytic activity (Regner et al, 2001). Cytolysis could occur

through the release of granules, perforin and granzymes from CTL towards target cells,

or via ligation of Fas receptor on target cells with Fas ligand on CTL. Both mechanisms

however have the same consequence; target cells will undergo apoptosis (Wang et al,

2004b). In vitro, CTL activity has been observed in mice infected with WNV and JEV

(Kesson et al, 1987; 1988; Murali-Krishna et al, 1994). In mice i.v. infected with low

dose of MVEV and WNV, although CD8+ T cells were induced in both infections

(Wang et al, 2003b; Licon Luna et al, 2002), further analysis showed that this subset of

T cells function differently even in a closely related flavivirus infections. The cytolytic

mechanism of T cells is involved in neuropathology of MVEV but in contrast; it

contributed to neuroprotection during WNV infection (Wang et al, 2004b; Licon Luna

et al, 2002). Mice deficient in either the granule exocytosis- or Fas-mediated pathways

of cytotoxicity or both were less susceptible to peripheral low dose MVEV infection,

demonstrating the immunopathological role played by CTL during virus infection.

Mullbacher and co-workers (2003) proposed that in wild type mice, CTL killed infected

endothelial cells lining the brain capillaries, causing a breakdown of the BBB and

allowing virus to enter the brain. However, in mice deficient in cytotoxicity pathways,

endothelial cells remained intact and did not undergo cytolysis and therefore BBB

breakdown and viral invasion of the CNS did not occur or was delayed.

1.3.3 SOLUBLE MEDIATORS

1.4.3.1 Cytokines

Cytokines are soluble proteins that have an important and key role in the induction and

maintenance of inflammation, the immune response and embryonal development (Kunzi

and Pitha, 2000). As potent inflammatory molecules, they have been implicated in many

CHAPTER 1

LITERATURE REVIEW

31

immune-mediated diseases (Lucey et al, 1996). The cytokine expression profile is

regulated by 2 different T-helper (Th) cell subsets (Kunzi and Pitha, 2000). It can either

be Th1 which is associated with production of IFNγ, IL-2, IL-12 and predominantly

leads to CMI; or Th2 which usually involves expression of IL-4, IL-5, IL-6 and IL-10

and associated with humoral mediated immunity and suppression of CMI (Lucey et al,

1996; Gouwy et al, 2005). The Th1 cells secrete factors that inhibit development of

cytokines from Th2 cells and vice versa.

The production of a particular subset of cytokines is dependent on many factors such

type of virus, route of inoculation, type of cells being infected as well as the genetic

background of the host. Infection of RNA viruses such as LCMV in rodent CNS

generated strong Th1 response with production of TNFα, IL-1, IL-6 and IFNγ observed

in the brain. In contrast, Sindbis virus that infects neurons and causes acute

encephalomyelitis was shown to elicit Th2 responses (reviewed in Kunzi and Pitha,

2000). However, in other virus-induced diseases, cytokine production cannot be

associated with particular Th cell subsets. In patients suffering from DHF during a 1996

DENV epidemic in India, severity of disease correlated with high levels mRNA and

serum IL-8 (Raghupathy et al., 1998). However, sera from Cuban patients suffering

from DF or DHF were shown to contain high levels of IL-10 compared to the

uninfected control group (Perez et al, 2004). Studies with JEV infection in humans

revealed that elevated levels of proinflammatory cytokines and chemokines are

associated with a poor prognosis (Winter et al, 2004). IFN, IL-6, IL-8 levels were high

in the cerebrospinal fluid (CSF) of JEV infected patients, which succumbed to disease

whereas RANTES/CCL5 chemokine was elevated in the plasma of the same patients.

However, whether the production of these soluble mediators contributes to the

pathogenesis of virus infection or simply as a result of viral burden is not known.

1.3.3.1.1 IFN type I

IFN type I consists of IFNα and IFNβ, and they are secreted by most cells in response to

infection or other stimuli. Murine IFN is encoded by a family of 11 genetically

closely related intronless genes while IFN is encoded by a single gene (Higashi et al,

1983; Shaw et al, 1983). The initial response occurs when virus enters the cell, and then

produces viral components including dsRNA. Presence of dsRNA activates

CHAPTER 1

LITERATURE REVIEW

32

transcription factor IRF-3, IRF-7, NF-kB and activating transcription factor (ATF2)/c-

Jun and subsequently inducing IFN production (Munoz-Jordan et al, 2003).

Once secreted, these cytokines act in both autocrine and paracrine manner by binding to

a cell surface receptor both on infected cells and neighbouring cells, activating the Janus

kinase (JAK), signal transducer and activator of transcription (STAT) and inducing

transcription of hundreds of IFN-inducible genes (Smith et al, 2005; Munoz-Jordan et

al, 2003; Haller et al, 2006). This cytokine acts within hours of infection and has 3

major roles in the cells: antiviral, antiproliferative and immunomodulatory (Nash and

Usherwood, 2000). Both in vitro and in vivo studies have shown the importance of this

cytokine in protecting infected cells or host from a fatal outcome during flavivirus

infections. Treatment of cell cultures with IFN type I prior to WNV and DENV virus

infection protects cells from death (Samuel and Diamond, 2005; Diamond et al, 2000).

During natural infection of flaviviruses, dendritic cells may be the first cell types to

produce IFN type I and initiate a cascade of antiviral reactions (reviewed in Navarro-

Sanchez et al, 2005; Chambers and Diamond, 2003). IFN may inhibit viral replication

by preventing translation and replication of infectious viral RNA via both RNase L and

Protein Kinase R dependent or independent mechanisms (Diamond and Harris 2001;

Diamond et al, 2000). Mice that are deficient in type I IFNs, or their receptor, show

increased susceptibility to flaviviruses (Lobigs et al, 2003a). In murine DENV-2

infection, IFNαβ has been demonstrated to be an important early immune mediator that

limits viral replication in extraneural tissues and subsequently reduces or prevents viral

spread to the CNS (Shrestha et al, 2004). Similar observations were also reported in

WNV infection (Samuel and Diamond, 2005). IFNαβ receptor deficient mice had 100%

mortality with a mean time to death of about 4 days post infection following

subcutaneous challenge of the virus. In contrast, wild type mice only had 62% mortality

and a longer mean time to death (Samuel and Diamond, 2005).

However, protection conferred by IFN type I apparently occurs only if cells or animals

are pretreated with the cytokine before infection. Treatments of IFN type I after

infection have very little or no effect on viral replication. WNV and DEN infections

have been demonstrated to inhibit the interferon signalling and its antiviral mechanism

(Guo et al, 2005; Munoz-Jordan et al, 2003). Treatment of cells or animals with IFNα

at 4 h after DENV or SLEV infection resulted in almost complete inability of IFNα to

induce antiviral activity (Diamond et al, 2000). Similarly, IFNα treatment of patients

CHAPTER 1

LITERATURE REVIEW

33

with JEV encephalitis had no significant effect on disease outcome (Solomon et al,

2003). This attenuation of the IFN antiviral effect is caused by the viral non-structural

proteins, NS4B and to a lesser extent NS2A and NS4A that block the activation of two

different interferon stimulate response element (ISRE) promoters in response to IFNβ as

shown by an in vitro study with DENV (Munoz-Jordan et al, 2003; reviewed in

Navarro-Sanchez et al, 2005). An amino acid substitution in the NS2A protein has been

shown to attenuate WNV NY strain’s ability to inhibit IFN type I induction, leading to

the loss of virulence in this flavivirus (Liu et al, 2006).

1.3.3.1.2 IFNγ

IFN is also known as IFN type II and the gene encoding this cytokine is mapped to

chromosome 12 and 10 in the human and mouse genome, respectively (Shtrichman and

Samuel, 2001). The expression of IFN-inducible genes is regulated through JAK-

STAT signal transduction pathway and some STAT-independent pathways (reviewed in

Samuel, 2001). This cytokine is produced by T and NK cells upon stimulation by

mitogenic and antigenic agents. IL-12 and IL-18, produced by macrophages and APCs,

are known to be the most potent IFN inducers (Boehm et al, 1997). Mice of different

genetic backgrounds have different capacities to produce IFN as reported for T cells

from C3H and C57BL/6 mice of being able to produce much higher IFN levels than

other mouse strains (Shtrichman and Samuel, 2001). IFN is the principal macrophage-

activating factor and it directly induces the synthesis of enzymes that mediate the

respiratory burst, allowing macrophages to kill phagocytosed microbes. In addition, this

cytokine also acts synergistically with TNF to induce production of nitric oxide. It also

increases the expression of MHC I and II and acts on B cells to promote switching to

the IgG2a and IgG3 subclasses and inhibits switching to IgG1 and IgE (Boehm et al,

1997). The protective nature of IFN has been demonstrated in the murine experimental

model with infections such as Hepatitis B virus, LCMV and mousepox virus

(Shtrichman and Samuel, 2001).

In flavivirus infection, early IFN expression produced by T cells was reported to

have a protective role and to contribute to the controlling and prevention of fatal

outcome in mice following WNV (New York isolate) infection (Wang et al, 2003a).

However, IFNγ does not seem to have any protective role during disease development

CHAPTER 1

LITERATURE REVIEW

34

following MVEV and YFV infections in mice (Lobigs et al, 2003a; Liu and Chambers,

2001). During i.v. infection with small doses of MVEV, IFN was reportedly to have no

beneficial role as mice deficient in IFN had only marginally increased mortality

compared to wild type mice (Lobigs et al, 2003a). Similarly, infection of YFV in IFN

knockout mice also did not increase the mice susceptibility to virus-induced encephalitis

although brain virus titres were higher in these mice indicating the role of IFN in

clearing the virus (Liu and Chambers, 2001). Interestingly, IFNγ has been demonstrated

to have an immunopathological role following i.p. infection with WNV Sarafend strain

(King et al, 2003). In IFNγ knockout mice, mortality reduced after WNV infection,

although kinetics of virus replication were similar in both wild type and IFNγ deficient

mice. In contrast, during DENV infection, IFN had dual roles; it elicited antiviral

activity against this virus but was also shown to help enhance virus infection that

eventually resulted in DHF (Sittisombut et al, 1995; Kontny et al, 1998; Libraty et al,

2001).

1.3.3.1.3 TNF

TNF is a 17-kDa proinflammatory cytokine, which is produced mainly by monocytes

and macrophages in response to infectious stimuli (Ravi et al, 1997). It is also a

pleiotropic soluble mediator as most all cells express at least one or the two types of

TNF receptors (TNFR I and TNFR II) (Darnay and Agarwal, 1999). While TNF I

transduces both death and survival signals, TNFR II only transduces survival signals

(Mizuno et al, 2001). TNF has an important role in various processes ranging from

promoting viral replication and evasion of host defences to activation of the immune

system by modulating the production of an array of cytokines (Ravi et al, 1997). Thus,

this cytokine could influence the outcome of diseases, by either being involved in

protection or exaggeration of virus infections. One of TNF inducers is IFN and these

two cytokines have been demonstrated to act together to provide antiviral activity and

protection during infections such as vaccinia virus (Sambhi et al, 1991), adenovirus

(Elkon et al, 1997) and herpes simplex virus (Kodukula et al, 1999). However,

induction of TNF in the CNS may also cause immunopathology as it is involved in

demyelination process, has cytotoxic effects on the endothelium and induces necrosis in

oligodendrocytes (Ravi et al, 1997)

CHAPTER 1

LITERATURE REVIEW

35

Production of TNF has been examined in several flavivirus infections. In vitro

exposure of a monocytic-like cell line to DENV resulted in high levels of TNF

production (Hober et al., 1996). TNF has been associated with increased permeability

of the BBB, which then permits virus to invade the CNS from the periphery. Upon

challenge with a lethal dose of WNV, TNF receptor deficient mice had a much higher

survival rate than those observed in wild type mice, demonstrating the role of TNF in

pathogenesis of WNV (Wang et al, 2004a). A similar pathogenic role of this cytokine

has been observed during i.c. challenge with JEV, whereby higher expression of TNF

mRNA was observed in the brain of infected mice (Suzuki et al, 2000). TNF levels

have also been examined in patients suffering from JE virus-induced encephalitis and

are rather linked to severity of disease than antiviral activity. The mean TNF levels in

serum and CSF of fatal cases of encephalitis were higher than in non-fatal cases,

indicating that TNF levels directly correlated with the mortality rate (Ravi et al, 1997).

It was revealed in these patients that serum TNF concentrations higher than 50 pg/mL

were associated with poor outcome of the disease.

1.3.3.2 Chemokines

Chemokines are low molecular weight chemotactic cytokines that bind to specific

G protein-coupled cell surface receptors (Thomsen et al, 2003). Chemokines are

responsible for regulating adhesive interactions with the vascular endothelium and

subsequently attracting leucocytes into the site of infection (Thomsen et al, 2003;

Asensio and Campbell, 2001). Thus, expression of chemokines will determine the

nature of inflammatory cells present at the infected tissues and consequently the types

of cytokines being produced. There is functional redundancy in the chemokine network

as one chemokine can bind to several receptors of the same family and one receptor can

bind to several related chemokines. They are either constitutively expressed or inducible

upon various stimuli and are classified into 4 groups based on the position of the first

cysteine; CC, CXC, CXC3 and C (Bajetto et al, 2002). The CC chemokines include

RANTES/CCL5, MIP1-/CCL3 and MIP-1/CCL4 and they induce chemotaxis of a

variety of cells including monocytes, CD8 and CD4 T cells (Bajetto et al, 2002). CXC

chemokines that include IP-10/CXCL10 induce migration of activated T and NK cells.

These chemokines are expressed in the CNS and could either be involved in

neuroprotection or neuropathogenesis in the brain.

CHAPTER 1

LITERATURE REVIEW

36

Generally, there is a typical profile of chemokines induced following virus infection,

which include RANTES, MIP-1, MIP-1, IP-10 and MCP-1 (reviewed in Thomsen

et al, 2003). Shirato and co-workers (2004b) compared expression of chemokines in

lethal (NY99) versus non-lethal WNV (Eg101) subcutaneous infection mice. Among

the CC and CXC chemokines that were under study, RANTES, MIP1, MIP-1 and IP-

10 mRNA expression were highly up-regulated upon lethal WNV infection which

suggested that these soluble mediators may be directly involved in the pathogenesis of

the virus. In contrast, BMAC mRNA was highly up-regulated later during infection in

non-lethal Eg101 compared to the lethal NY99 WNV virus. BMAC, which is

chemotactic agent for B cells and monocytes, may contribute to the protective nature of

the immune system later in infection by inducing macrophages migration (Shirato et al,

2004b). RANTES/CCL5 chemokine has been demonstrated to have a more protective

than immunopathological role upon infection with WNV strain NY99. Infection of mice

deficient in the receptor for RANTES/CCL5 (CCR5) resulted in higher mortality

(100%) as well as increased viral burden and reduced leucocytic infiltration when

compared to the wild type mice (Glass et al, 2005).

1.4 GENETIC RESISTANCE TO FLAVIVIRUSES

Host genetic resistance to certain pathogens is a very important factor in determining

the severity of virus-induced diseases. The susceptibility or resistance to disease varies

between individuals upon exposure to viral pathogens and this trait can be passed on to

the next generation (Brinton, 2001). At present, there are already a number of genes

identified to confer resistance to viral-induced diseases in the infected host (Brinton,

1997). Some operate independently while others may be synergised with other

components of the immune system to mediate an effect (Urosevic et al, 2000). The most

important features of host natural resistance to viruses are: it usually acts early during

the course of infection, it lacks the unique specificity of the immune system and it

confers protection only against a specific group or family of viruses (Urosevic and

Shellam, 2002). Resistance to influenza virus in mice for example is attributed to the

Mx gene (Haller et al, 1979). This resistance gene is mapped to chromosome 16, is IFN-

inducible and reduces viral replication at the level of viral mRNA synthesis (Krug et al,

1985). Another example is the Cmv1 gene, which restricts murine cytomegalovirus

CHAPTER 1

LITERATURE REVIEW

37

infection in the spleen (Scalzo, 1990). This gene is mapped to murine chromosome 6

and contributes to host resistance through NK cells (Scalzo et al, 1995). However, the

most relevant for this study is the Flvr gene conferring resistance to Flavivirus infection

as will be described in the following sections.

1.4.1 FLAVIVIRUS RESISTANCE IN HUMANS

Evidence of innate resistance to flaviviruses in humans is indirect at present. In Africa,

where YF was highly endemic, there were groups of people who seemed to be resistant

to the virus and did not suffer from any clinical manifestation of the disease whereas

strangers coming to this area were highly susceptible (Sabin, 1954). The mechanism of

human resistance is unknown but it’s been suggested that thousands of years of

exposure to YFV has eliminated individuals carrying susceptibility genes, while those

having the resistance genes survived the infection, resulting in genetically determined

natural human resistance to YFV (Sabin, 1954). By contrast, the shorter period of

exposure in the South American Indians to the same virus resulted in the mixed

incidence of severe and mild YF among these people (Sabin, 1952b).

1.4.2 FLAVIVIRUS RESISTANCE IN MURINE MODELS

1.4.2.1 History and development

The phenomenon of host innate resistance to flaviviruses is well established in murine

models. In 1931, Sawyer and Lloyd reported a variation in survival seen in different

mouse strains following i.c. inoculation of YFV. Lynch and Hughes (1936) then

demonstrated a strain of mice from the Rockefeller Institute in New York (Det) were

also less susceptible to i.c. challenge to YFV infection. Following this, Webster

(Webster and Clow, 1936; Webster, 1937) created four mouse strains with different

susceptibility to Salmonella enteritidis and two flaviviruses, louping ill (LI) and St.

Louis encephalitis (SLE). These mouse strains were known as bacteria-susceptible-

virus-susceptible (BSVS), bacteria-resistant-virus-susceptible (BRVS), bacteria-

susceptible-virus-resistant (BSVR) and bacteria-resistant-virus-resistant (BRVR).

Studies using these mouse strains showed that resistance to bacteria and to virus are

inherited independently. The brain viral titres in virus-susceptible strains were found to

be higher than in virus-resistant strains (Webster and Clow, 1936; Webster, 1937).

CHAPTER 1

LITERATURE REVIEW

38

Sabin made a further discovery of murine flavivirus resistance in 1944 when he

demonstrated that an outbred strain of mice at the Princeton Rockefeller Institute,

known as PRI mice were 100% resistant to i.c. challenge of YFV. Brain viral titres in

PRI mice were found to be 10,000 to 100,000 fold less than those found in susceptible

Swiss mice (Sabin, 1952a). Studies with other viruses showed that this resistance is

flavivirus specific as PRI mice were also resistant to WNV, JEV, SLEV, DENV and

Russian Spring Summer encephalitis virus (Sabin, 1952a; 1952b; 1954). The flavivirus

resistant mice however are equally susceptible to other arboviruses including Sindbis

virus, Ross River virus and Chikungunya virus (Hanson et al, 1969; Shellam et al,

1998). Sabin’s studies also suggested that this flavivirus-resistance-gene is inherited as

an autosomal, dominant trait controlled by alleles of a single genetic locus, which is

later known as Flv (Sabin, 1952b, Green 1989).

1.4.2.2 Flavivirus resistance in wild mice

Genetically controlled flavivirus resistance is not a common feature in laboratory strains

and it has only been previously described in a few mouse strains such as in Det, BSVR,

BRVR and PRI mice. On the other hand, resistance was shown to be widespread and

abundant in wild mice (Brinton and Perelygin, 2003). M. m domesticus mice trapped in

Maryland, Virginia and California in America in the early 1970s showed resistance to

17D YFV challenge (reviewed in Brinton and Perelygin, 2003). Additionally, wild

M. m domesticus mice found in Australia were also resistant to i.c. infection with

MVEV (Sangster and Shellam, 1986). Resistance in these wild mice appeared to be

attributed to the Flv gene (Sangster et al, 1998).

In addition, inbred strains derived from wild mice within the Mus complex have also

been tested for their flavivirus resistance. CASA/Rk and CAST/Ei strains derived from

M. musculus castaneus revealed similar resistance to C3H/RV (RV) mice. In contrast,

MOLD/Rk strain derived from M. musculus molossinus has different a resistance

phenotype than RV mice. MOLD/Rk mice were shown to have only moderate or

intermediate resistance to flaviviruses (Sangster et al, 1993). These mice survived i.c.

infection with 17D YFV but not with MVEV. It has been suggested that the occurrence

of flavivirus resistance in wild mice is due to the selective pressure of the virus

infections (Brinton, 1981).

CHAPTER 1

LITERATURE REVIEW

39

1.4.2.3 Development of congenic flavivirus mouse resistant strains

In order to study the resistance conferred by Flv possible under controlled laboratory

conditions, a virus-resistant mouse strain congenic to inbred-virus susceptible mice was

developed (Groschel and Koprowski, 1965). The first congenic strain established was

C3H.RV (RV) (later known also as C3H/PRI-Flvr), created by 8 generations of

backcross breeding between PRI and C3H/He mice (Goodman and Koprowski, 1962a;

Groschel and Koproswski, 1965). For many years, this was the only flavivirus resistant

congenic strain available until new strains were developed in the 1990’s. C3H/MOLD-

Flvmr

(MOLD) was generated by introducing the genetic locus from MOLD/Rk strain

into the genetic background of C3H/HeJARC (HeJARC) mice; by backcross breeding

and further brother and sister matings (Urosevic et al, 1999). MOLD/Rk mice showed

different resistance phenotype than PRI mice, where the former mice were only resistant

to i.c. infection of YFV but not MVEV OR2 (minor resistance) (Sangster et al, 1993).

Thus, YFV virus was used to select the offspring from the backcross breeding carrying

the moderate resistance gene. By limited allelism analysis, the minor resistance

expressed in MOLD/Rk mice were shown to be an allelic variant at the Flv locus and

thus has been designated as Flvmr

(Sangster et al, 1993). Another resistant strain was

also created in the same laboratory using a similar approach, this time using resistant

wild Mus domesticus mice trapped in Dubbo, New South Wales as the source of

resistance gene. This new strain was called C3H.M.domesticus.Flvr-like (DUB) and was

created by 11 generations of backcrossing to HeJARC mice (Urosevic et al, 1999). In

vivo and in vitro studies showed that DUB mice demonstrated similar but slightly

stronger levels of resistance than RV mice (Silvia et al, 2001). The susceptible HeJARC

mice used to create this congenic mouse strain were shown to be a lipopolysaccharide-

responsive subline of C3H.HeJ (HeJ) mice (Silvia and Urosevic, 1999). Urosevic and

co-workers (1999) reported that these three congenic resistant mouse strains (RV,

MOLD and DUB) carry different segments of the chromosome 5, encompassing Flv

embedded within the same genetic background of flavivirus-susceptible C3H mice. The

sizes of donor-derived chromosomal regions flanking the selected resistance alleles are

estimated to be 9 and 11cM in DUB and MOLD mice, respectively (Urosevic et al,

1999).

CHAPTER 1

LITERATURE REVIEW

40

1.4.2.4 Resistance expression in mice

Flavivirus resistant mice are only resistant to diseases induced by flaviviruses but they

can still be infected by the viruses. Thus, virus can still grow in the cells or tissues of the

resistant animals although at a much lower titres than in susceptible mice. Webster and

Clow (1936) were the first to provide data on virus growth in susceptible HeJ and

resistant RV mice. They showed that during SLEV infection, viral load in the brain of

infected resistant mice remained lower than in susceptible mice. A similar growth

pattern was also observed during i.c. challenge with WNV in the same strain of mice

(Hanson et al, 1969). Viral titres rose rapidly in susceptible mice whereas the virus

growth was delayed in resistant mice with maximum titres at about 3 log lower than in

susceptible mice.

Viral replication in brains of other resistant mouse strains has been studied by Urosevic

and co-workers (1999). Intracerebral challenge of MVEV OR2 in HeJ mice resulted in

rapid virus growth and reached maximum on day 4 p.i. and mice died two days later. In

mild resistant MOLD mice, viral titres were significantly lower than in susceptible mice

although mice eventually succumbed to the infection. In resistant RV and DUB mice

however, virus replicated to a much lower level in comparison to susceptible and

moderately resistant mice. By day 10 p.i., no virus was detected and mice survived the

infection. YFV challenge was lethal for susceptible mice while all resistant mouse

strains were equally resistant to this virus (Urosevic et al, 1999).

1.4.2.5 Resistance expression in cell culture

Since the flavivirus resistance gene does not have any effect on viral entry or initial

viral replication, similar numbers of resistant and susceptible derived mouse embryo

fibroblasts (MEF) were shown to be infected with WNV by immunofluorescence

(Brinton-Darnell and Koprowski, 1974). Similar numbers of thioglycollate-elicited

macrophages from both resistant and susceptible mice were also found to be initially

infected with WNV (Silvia, 1999). However, virus replication is still lower in cells or

tissues derived from resistant mice in comparison to susceptible mice indicating that

innate resistance to flavivirus is expressed at the level of the individual cells. Ten to

hundred fold lower WNV titres were produced in MEF from resistant mice, compared

to MEFs from susceptible mice (Briton-Darnell and Koprowski, 1974). Nevertheless the

CHAPTER 1

LITERATURE REVIEW

41

maximum difference in viral titres between resistant and susceptible mice in vitro is not

as great as in vivo. For example, infection of resistant peritoneal macrophages with

MVEV OR2 resulted in only 10 times titres lower replication than in susceptible cell

cultures whereas the difference in maximum brain titres was more than 103.5

TCID50

units (Silvia et al, 2001). Expression of resistance has been found in primary brain cell

cultures, spleen cells, kidney cells, peritoneal macrophages and mouse-embryo

fibroblasts (Webster and Johnsen, 1941; Goodman and Koprowski, 1962a; Vainio

1963a; Vainio, 1963b; Hanson et al, 1969; Silvia et al, 2001; Pantelic et al, 2005).

However, primary cultures derived from astrocytes and skin fibroblasts do not

conclusively express flavivirus resistance in vitro (Silvia, 1999).

1.4.3 THE MECHANISM OF FLAVIVIRUS RESISTANCE

The action of the gene Flvr is virus specific; it only provides resistance to flavivirus

infections but not to other viruses (Sabin, 1952b; Shellam et al, 1988). Among the

flaviviruses that have been tested are WNV, KUNV, Alfuy virus, Kokobera virus,

SLEV, DENV, MVEV and Russian spring-summer encephalitis virus (reviewed in

Shellam et al, 1988). However the resistance conferred by the Flvr gene does not protect

the animal from non-flaviviruses such as arenaviruses, lymphocytic choriomeningitis,

picornavirus, poliovirus, Western equine encephalitis, Semliki Forest virus and others

(reviewed in Brinton and Perelygin, 2003). The major phenotypic features of the Flv

resistance are decreased production of infectious virus and limited spread to

surrounding cells (Urosevic and Shellam, 2002). From in vitro studies, it was shown

that the resistance conferred by Flvr gene acts intracellularly at the level of virus RNA

synthesis and assembly rather than at the level of virus entry (Brinton, 1983).

Defective interfering (DI) viruses were suggested to play a role in Flvr mediated

resistance. These DI particles usually contain deletions, often due to inversions or

rearrangement of the gene (reviewed by Urosevic and Shellam, 2002). Because

defective interfering (DI) particles do not produce replicase, they bind to the standard

virus replicase and as a consequence interfere with the infectious viral replication. DI

particles replicate to higher levels at high MOI when all infected cells contain infectious

viral genomes, resulting in reduced production of standard virus progeny (Dimmock and

Primrose, 1989; Dulbecco, 1990). However, evidence obtained so far suggested that

production of DI viruses is rather associated with persistently infected cell culture and is

CHAPTER 1

LITERATURE REVIEW

42

quite uncommon in animal studies. Persistently infected MEF derived from resistant

mice were shown to produce more DI and less infectious virus than cells from

susceptible mice upon WN infection (Brinton, 1983). Additionally, DI particles were

also observed during infection with Banzi, MVE and JE viruses (reviewed in Urosevic

and Shellam, 2002). Following challenge with Banzi virus, DI was only found in brains

of resistant mice but not in susceptible during i.p. infection but could not be

demonstrated from brains of both strains upon i.c. infection (Smith, 1981). When both

susceptible and resistant mice were infected i.c. with MVE, no accumulation of DI was

observed in resistant mice as well (Urosevic et al, 1997a). Thus, since the production of

DI particles was not always consistent with the resistance phenotype, DI viruses

possibly do not play a major role in Flvr mediated resistance but it may reflect the

characteristics of cell culture under study as well as the strain of virus used to infect the

mice.

The role of IFN type I in flavivirus resistance has not been found to be critical for its

expression (Vainio et al, 1961; Urosevic and Shellam, 2002). This is in contrast to the

Mx gene which confers resistance to influenza virus-induced diseases and it requires

induction of IFN for its induction (Haller et al, 1980). Resistance in RV mice was not

abrogated during i.c. 17D YFV infection when the antiviral effect of IFN was

neutralised by sheep anti-IFN αβ antibodies (Brinton et al, 1982). Brain viral titres

were slightly higher in IFN-depleted resistant mice but were still significantly lower

than in susceptible mice. Additionally, IFN type I production in brains of infected

susceptible mice was higher than in resistant mice following i.c. infection with KUN

and WNV (Shueb, 2002; Hanson et al, 1962). IFN production in these animals

correlated directly with viral titres and showed that Flv mediated resistance was indeed

IFN independent.

In contrast, RV derived MEFs and macrophages showed higher sensitivity to the

antiviral effect of IFN than HeJ derived MEFs (Hanson et al, 1969; Pantelic et al,

2005). Addition of exogenous IFN before WNV infection produced several times lower

viral titres in resistant than in susceptible cell cultures (Hanson et al, 1969; Pantelic et

al, 2005). Thus, although Flvr involvement in the reduction of viral replication is IFN

independent, it appears that IFN has the ability to potentiate this resistance effect. This

is possibly mediated by IFN-inducible factors encoded at loci closely linked to Flvr and

expressed only upon IFN stimulation (Urosevic and Shellam, 2002).

CHAPTER 1

LITERATURE REVIEW

43

NO is another antiviral molecule that has been implicated in many biological processes

including host defence against intracellular pathogens (Reiss and Komatsu, 1998;

Nathan, 1992). As NO was shown to contribute resistance to murine Hepatitis virus

(Pope et al, 1998) and since the Flv locus was mapped to the same region as NOS-1, a

possibility existed that the NOS-1 could be a potential candidate for the gene that

confers resistance to flaviviruses. However, Silvia and co-workers (2001) monitored the

level of brain NO production in susceptible HeJARC, RV and DUB mice upon i.c.

challenge with MVEV OR2 and found no difference in NO brain tissue levels before or

after the infection. Similar results were demonstrated when these 3 mouse strains were

infected i.c. with KUNV MRM 16 (Shueb, 2002). In vitro studies using mouse

macrophages infected with WN virus also showed similar levels of NO production in

these cells derived from either resistant or susceptible strains (Silvia et al, 2001). These

studies thus indicated that murine innate resistance to flavivirus conferred by Flvr is NO

independent.

1.4.4 ANALYSIS OF GENE CANDIDATE FOR FLAVIVIRUS RESISTANCE

GENE

Although the phenotypic expression of Flv has been well characterised in vivo and in

vitro, the search and validation for the possible gene candidate for Flv are still in

progress. Using a three-point backcross linkage analysis, Flv locus was initially mapped

to mouse chromosome 5, between the retinal degradation (rd) locus and the

glucoronidase structural gene (Gus-s) in the region encompassing 23cM of genetic

distance (Shellam et al, 1993; Urosevic et al, 1993; Sangster et al, 1994). However, by

employing a low-resolution genetic mapping, this region was narrowed to 1 cM

(Urosevic et al, 1995) and then further narrowed to 0.45cM using high-resolution

microsatellite analysis (Urosevic et al, 1997b).

However, using these genetic maps and a positional cloning approach, the Oas1b gene

has been recently identified as a possible Flv gene candidate (Perelygin et al, 2002;

Mashimo et al, 2002). The premature stop codon in the fourth exon of the Oas1b gene

present only in susceptible but not in resistant mice. This may result in the expression of

proteins that lack 30% of the C-terminal sequence compared to the proteins that are

expressed in resistance mice. This nonsense mutation results in a predicted truncated

CHAPTER 1

LITERATURE REVIEW

44

protein lacking a catalytic domain in susceptible hosts (Lucas et al., 2003). The intact

Oas1b protein is expressed in resistant mice including RV, BRVR, CASA/Rk and

CAST/Ei while similar predicted truncated Oas1b protein should be expressed in

susceptible BALB/c, HeJ, C57BL/6 and CBA/J, based on gene sequence analysis

(Brinton and Perelygin 2003; Mashimo et al, 2002). Oas1b expression in transfected

MEF has been shown to provide some protection against WNV infection (Perelygin et

al, 2002). Additionally, constitutive expression of Oas1b in stable mouse neuroblastoma

cells limited cell-to-cell spread of WNV (Lucas et al, 2001). Kajaste-Rudjitski and co-

workers (2006) showed that the Oas1b gene provides antiviral protection against WNV

by limiting the accumulation of positive-stranded viral RNA in infected cells. Urosevic

and co-workers (1997a) also reported similar observation. Viral dsRNA replicative form

was found to be at greater levels in brains of resistant mice upon i.c. challenge with

MVEV than in brains of susceptible mice (Urosevic et al, 1997a).

To date, the only well established biochemical function of Oas1b gene is the activation

of a ubiquitous 2-5A dependent, single strand specific, cytoplasmic endoribonuclease,

RNase L. (Zhou et al, 1993). However, recent study indicates that although RNase L

contributes to the cellular antiviral activity against flaviviruses, RNase L activation is

not part of Oas1b-mediated flavivirus resistance phenotype (Kajaste-Runitski, 2006).

Thus, although the flavivirus resistant gene has been identified, the mechanism by

which it regulates the levels of viral RNA in infected cells and confers protection

against fatal encephalitis remains to be elucidated. Further study is also required to

validate the Oas1b gene in innate immunity against other flaviviruses since most studies

involving this gene were conducted during infection with WNV only.

1.4.5 FACTORS INFLUENCING THE HOST INNATE RESISTANCE TO

FLAVIVIRUSES

Although Flvr gene confers resistance to flaviviruses in mice, protection in many

instances is not complete and infection of flaviviruses can lead to fatal outcomes. This

depends on several factors such as age of mice, immune status, virus strain and dose or

route of inoculation (Sabin, 1952a; Goodman and Koprowski, 1962; Hanson et al,

1969). Newborn RV mice (1-2 days old) succumbed to i.c. infection of YFV strain 17D

while suckling mice behaved like adult mice and survived the infection (Sabin 1952b).

In addition, upon flavivirus infection in 7-day-old suckling BRVR and BSVS mice,

CHAPTER 1

LITERATURE REVIEW

45

both mouse strains eventually died but resistant mice succumbed to the infection 2 days

later than susceptible mice (Goodman and Koprowski, 1962a). The levels of replicating

virus in these dying resistant mice however was lower than in susceptible mice,

indicating that the Flvr gene is expressed from a very early age to restrict virus growth.

Death observed in this group of mice may be associated with an immature immune and

central nervous systems (Urosevic, 2000). Sabin also demonstrated the effect of

different strains of flaviviruses on the survival of resistant mice. While RV adult mice

exhibited complete resistance to YFV of strain 17D, 24% of them died from French

neurotropic strain of YFV (Sabin, 1954). Large doses of virulent viruses were also

shown to produce high mortality in resistant mice, although the onset of disease

symptoms and time of death usually were still delayed in comparison to susceptible

mice (Goodman and Koprowski, 1962a; Jacoby and Bhatt, 1976).

Flv gene only reduces viral replication and eventually viral spread to other cells or

tissues during infections. However, complete clearance of virus is pivotal to prevent

death of the infected resistant host (Bhatt and Jacoby, 1976). Thus, the Flv gene works

concurrently with a competent immune system to confer protection against a fatal

disease outcome. Both humoral and cell-mediated immunity are vital for virus clearance

and protection against fatal flavivirus infection in flavivirus resistant mice. Indeed,

immunosuppression has been shown to reduce the host innate resistance to flaviviruses

(Goodman and Koprowski, 1962a).

Camenga and co-workers (1974) provided evidence for the importance of antibodies in

the recovery of flavivirus infection. Cyclophosphamide impairs the antibody response

and resistant animals treated with this drug became susceptible to sublethal infection of

WNV. The requirement for humoral immunity was also demonstrated during i.p. Banzi

infection in RV mice. RV mice are highly resistant to i.p. Banzi infection and

cyclophosphamide treatment rendered these mice susceptible to Banzi challenge (Bhatt

and Jacoby, 1976). The requirement for CMI was demonstrated when thymectomised

RV mice, which have an impaired T cell response but are still capable of producing

antibody, became susceptible to Banzi infection (Jacoby et al, 1980). However,

although these immunosuppressed resistant mice were susceptible to flavivirus

infection, they exhibited slower virus spread; lower virus titres as well as delayed time

to death compared to susceptible mice that received the same treatment. This result

indicated that although functional host immune system is required for the complete

CHAPTER 1

LITERATURE REVIEW

46

clearance of the virus and survival of the animal, both humoral and CMI do not

contribute to the Flv resistance mechanism (Brinton and Perelygin, 2003).

Virus strain is also important in determining the outcome of disease after flavivirus

infection. Intracerebral infection of YFV strain 17D caused high mortality in susceptible

HeJ mice only, while MVEV OR2 infection resulted in mortality in both susceptible

and minor resistant MOLD mice but not highly resistant RV and DUB mice (Urosevic,

et al, 1999; Shueb et al, 2005). In contrast, all susceptible, minor resistant and highly

resistant mice succumbed to KUNV MRM16 infection, although the time to death was

delayed and brain viral titres were much lower in resistant mice in comparison to

susceptible mice (Shueb et al, 2005).

1.5 AIMS

Flavivirus infections cause diseases ranging from febrile illnesses to encephalitis and

haemorrhagic fever. In mice, infection with these viruses causes encephalitis and thus,

mice have been used as an experimental model to study flavivirus-induced encephalitis

in humans. Severity of the diseases is influenced by many complex factors such as the

type of infecting virus, virus dose, route of inoculation, immune status of the host and

host genetic resistance to the virus.

There are currently several congenic inbred mouse strains which are either susceptible

or resistant to flavivirus infections. These mouse strains have been used extensively to

study a genetic basis of flavivirus resistance in mice. A single autosomal genetic locus,

flavivirus resistance locus (Flv) was shown to control inborn resistance to flaviviruses in

mice (reviewed in Brinton and Perelygin, 2003). This inherited resistance gene confers

protection early in flavivirus infection by reducing the viral titres. Flv was recently

identified by positional cloning to be the OAS 1b gene, although the mode of its action

is still not known (Mashimo et al, 2002; Perelygin et al, 2002). It has been shown in our

laboratory that Flv-controlled resistance cannot provide complete protection against all

flavivirus infections (Shueb et al, 2005). Initial studies performed in this laboratory

demonstrated that upon i.c. challenge with WNV Sarafend or KUNV MRM16, resistant

mice succumbed to the infection. In contrast, i.c. infection with MVEV OR2 did not

cause any disease or death to resistant mice despite its great lethal effect in susceptible

CHAPTER 1

LITERATURE REVIEW

47

mice. On the basis of this finding, it was assumed that the virulence of flaviviruses in

resistant mice is different from their virulence in susceptible mice. In addition, previous

findings in this laboratory also suggested that development of fatal encephalitis in

resistant DUB mice during intracerebral infection with some flaviviruses did not

necessarily coincides with high levels of virus replication and that Flv-controlled

resistance, although operative, was not sufficient to protect flavivirus resistant mice

from developing lethal diseases. Thus, the general aim of this project was to further

characterise the response of resistant and susceptible host to infection with the same

viruses and to identify host factors that contribute to either the recovery or morbidity of

resistant mice following infection with different but closely related flaviviruses. We

hypothesise that additional virulence factors, other than virus ability to replicate at high

levels, may become important to resistant host in influencing the host immune response

and underlying pathogenesis of flavivirus infection.

The first aim of this study was to characterise the virulence of 3 different flaviviruses;

KUNV MRM16, MVEV OR2 and WNV Sarafend in susceptible and resistant mice,

and to determine the effect of alteration of host defence mechanisms on virus virulence

and outcome of infection. Intracerebral, intranasal (i.n) and i.p. challenges of the viruses

were performed to determine the neurovirulence and neuroinvasiveness of KUNV,

MVEV and WNV. Since mice maybe resistant to i.p. challenge, studies were

undertaken to examine the role of three host defence mechanisms; the blood brain

barrier, macrophages and T cells, in the outcome of virus infection following i.p.

challenge. BBB permeability was altered using SDS and LPS in mice during i.p.

infection of the viruses. BBB is responsible for restricting the influx of foreign

molecules into the brain and its breaching has been reported to increase mice

susceptibility to virus infection (reviewed in Chambers and Diamond, 2003).

Additionally, thioglycollate, clodronate and T cells depletion were used to study the

influence of macrophages and T cells in severity of flavivirus infection.

The second aim was to characterise in vitro infection of KUNV, MVEV and WNV. In

addition to standard laboratory cell lines such as Vero cells, primary cells including

thioglycollate-elicited macrophages isolated from susceptible HeJ and resistant DUB

mice as well as dendritic cells isolated from susceptible C57BL/6 mice were used in this

study. Extensive studies have been performed previously in this laboratory, which

demonstrated that macrophages could be infected with WNV in vitro. However, parallel

CHAPTER 1

LITERATURE REVIEW

48

and comparison analysis of different flavivirus infections in cell cultures have never

been performed before and therefore, in this study, the aim was to investigate variations

in viral titres, cytopathic effect (CPE) as well as in cytokine productions following

KUNV, MVEV and WNV infection. This would provide additional insight into the

virus characteristics that may assist in better understanding of their different virulence

observed in vivo.

The third aim of this project was to examine MVEV and KUNV pathogenesis in

susceptible and resistant mice during i.c. infection and to determine factors that could be

attributed to the different outcome of infection following these 2 flavivirus infections.

The pathogenesis of these flaviviruses was studied and compared by examining brain

viral titres and characterising the types of infiltrating leucocytes in the CNS. In addition,

cytokine production was examined to study the correlation between the induction of

certain cytokines, virus clearance and severity of disease. In order to investigate the role

of T cells in virus pathogenesis or host recovery, depletion of these cells was performed

in mice infected with KUNV or MVEV, and mortality was monitored.

Studies conducted by other investigators on pathogenesis of flaviviruses were mainly

performed in flavivirus susceptible animals and cells. However, there were a limited

number of similar studies undertaken in flavivirus resistant hosts. In this regard, this

project will shed further light on flavivirus infection in resistant hosts and cells and

provide insight into the pathogenic mechanisms involved in flavivirus susceptible and

resistant mice.

CHAPTER 2

MATERIALS

49

2.0 CHAPTER 2: MATERIALS

2.1. REAGENTS

Acetic acid, glacial May and Baker, Australia (M & B)

Actinomycin D Sigma-Aldrich, USA (Sigma)

Ammonium chloride (NH4Cl) BDH Laboratory Supplies, England

(BDH)

Benzyl penicillin Commonwealth Serum Laboratories,

Australia (CSL)

Calcium chloride (CaCl2) BDH

Crystal violet Sigma

DePeX BDH

3, 3’-Diaminobenzidine tetrahydrochloride

(DAB) with Metal enhancer tablet set Sigma

Dimethyl sulphoxide (DMSO) BDH

Disodium hydrogen orthophosphate

(Na2HPO4) BDH

DNase 1 Boehringer Manheimann, Germany

Ethanol Merck Pty Limited, Australia

(MERCK)

Ethylenediamine tetra-acetic acid (EDTA)

BDH

Foetal calf serum Commonwealth Serum Laboratories,

Australia (CSL)

Formaldehyde MERCK

Gentamycin Delta West Ltd, Australia

Goat anti-rat anti-CD4 PE conjugated Pharmingen

Goat anti-rat anti-CD8 PE conjugated Pharmingen

CHAPTER 2

MATERIALS

50

Goat anti-rat anti-CD11b PECy5 conjugated

Pharmingen

Goat anti-rat anti-CD19 PE conjugated Pharmingen

Heparin Sigma

HEPES Buffer Gibco, USA

Horse serum donor herd Gibco

Hydrochloric acid (HCl) BDH

Hydrogen peroxide (H2O2) BDH

Lipopolysaccharide E.coli serotype

0128:B12 Sigma

Lycopersicum esculentum (tomato lectin) Vector Laboratories Inc, USA

L-glutamine Sigma

Magnesium chloride (MgCl2) BDH

2β-Mercaptoethanol Sigma

Methanol BDH

Methylene blue BDH

Mouse anti-mouse H-2Kk monoclonal

antibodies (MHC I) FITC conjugated Pharmingen

Mouse anti-mouse I-Ek monoclonal

antibodies (MHC II) FITC conjugated Pharmingen

New born calf serum (NCS) CSL

Paraformaldehyde Prolabo, Paris

Percoll Amersham Biosciences, Sweeden

Phosphoric acid (H3PO4) BDH

Potassium bicarbonate BDH

CHAPTER 2

MATERIALS

51

Potassium cacodylate Promega

Potassium chloride (KCl) BDH

Potassium dihydrogen orthophosphate

(KH2PO4) BDH

Propidium iodide Pharmingen

Protease inhibitor tablet Roche

Proteinase K Sigma

READY-SET-GO! Mouse Th1/Th2 ELISA Ebiosciences

RPMI 1640 Gibco

Sodium chloride (NaCl) BDH

Sodium dodecyl sulphate (SDS) BDH

Sodium hydrogen carbonate (NaHCO3) BDH

Sodium hydroxide (NaOH) BDH

di-Sodium hydrogen orthophosphate

(Na2HPO4) BDH

Sodium pyruvate BDH

Streptavidin-horseradish peroxidase DAKO

Thioglycollate Difco Laboratories, USA (Difco)

Tissue-tek O.C.T. Compound Sakura Finetek, USA

READY-SET-GO TNF ELISA Ebiosciences

TRIS Gibco

TRIS-HCl Sigma

Trypan blue Difco

CHAPTER 2

MATERIALS

52

Trypsin Difco

Tween-20 BDH

Xylene MERCK

2.2 CELL CULTURE MATERIALS

6-well culture plates Falcon Becton Dickinson (Falcon)

24-well culture plates Falcon

96 well culture plates Falcon

15 mL polystyrene conical tube Falcon

50 mL polypropylene tubes Falcon

1.5 mL polypropylene tubes Sarsredt, Numbrecht, Germany

2 mL polypropylene tubes Nuclon, USA

1.8 mL cryotubes Nunclon,

225 cm2 cell culture flask Costar, Corning Incorporation, USA

75 cm2 cell culture flask

Nunclon

Teflon well inserts Savillex Corporation, St Paul, USA

26-G needle Becton and Dickinson

CHAPTER 2

MATERIALS

53

2.3 BUFFERS, SOLUTIONS AND MEDIA

2.3.1 CELL STUDIES

All reagents were made up in double distilled water (DDW) and stored at room

temperature (25oC), unless otherwise stated. Where necessary, the solutions were

adjusted to the required pH with concentrated HCl or concentrated NaOH.

2.3.1.1 Growth media

RPMI 1640 medium was used for cultivation of Vero cells, L929 cells, primary cultures

of mouse macrophages, primary splenocytes cultures, brain mononuclear cells and

hybridoma cell lines YTS 169 and YTS 191. Medium was warmed shortly before use

with FCS or NCS was added as required: 10% (v/v) for cell growth and 2% (v/v) for

virus growth. For cultivation of hyridoma cell lines, 2-mercaptoethanol and sodium

pyruvate were also added.

RPMI 1640 liquid

Supplied pre-made by Gibco BRL

*L-glutamine 0.29 g/L

Benzyl penicillin 100 mg/L

Gentamycin 10 g/L

* L-glutamine was added to the medium shortly before use. FCS was added as required

and the medium was stored at 4oC.

RPMI 1640 powder

RPMI pre-made powder 110 g

NaHCO3 20 g

Benzyl penicillin 100 mg/L

Gentamycin 10 g/L

*L-glutamine 0.29 g/L

pH of the medium was adjusted to 7.4, sterilised by pressure filtration and stored at 4oC.

* L-glutamine was added to the medium shortly before use. NCS was added as

required.

CHAPTER 2

MATERIALS

54

2.3.1.2 Cell culture solutions

0.1 M L-Glutamine stocks

L-glutamine 0.1 M

The solution was filter sterilised through 0.22 μm filters into 10 mL aliquots and stored

at –20oC.

Phosphate Buffered Saline (PBS)

NaCl 8.0 g/L

KCl 0.2 g/L

Na2HPO4 0.91 g/L

KH2PO4 0.12 g/L

pH was adjusted to 7.4, autoclaved for sterilisation and stored at 4oC.

Stock EDTA (0.4%)

EDTA 4 g/L

The solution was filter sterilised through 0.22 μm filters, dispensed in 10mL aliquots

and stored at –20oC

Stock Trypsin (2%)

Trypsin 20 g/L

The solution was filter sterilised through 0.22 μm, dispensed in 5mL aliquots and stored

at –20oC.

PBS/Trypsin/EDTA

Trypsin 0.5 g/L

EDTA 0.2 g/L

The reagents were dissolved in PBS and stored at 4oC

100x Sodium Pyruvate (100 mM)

Sodium pyruvate 1.1 g

Double distilled water 0.1 L

The solution was filter sterilised through 0.22 μm, dispensed in 10 mL aliquots and

stored at –20 oC.

CHAPTER 2

MATERIALS

55

1000x 2-Mercaptoethanol (50 mM)

2-Mercaptoethanol 50 mM

Solution was made in sterile distilled water and stored at 4oC.

Methylene Blue – Formaldehyde Stain

Methylene blue 10 g/L

Formaldehyde 0.1 L

Methylene blue was added into 900 mL distilled water and stirred overnight. The

solution was then filtered through Whatman number 1 film paper and added with

formaldehyde.

Saturated Ammonium Sulphate

Ammonium sulphate 761 g

Distilled water 1 L

Ammonium sulphate was added to distilled water and stirred slowly. The pH was

adjusted to 7.0 and the solution was autoclaved.

2.3.2 IMMUNOHISTOCHEMISTRY

10xTris Buffered Saline (TBS)

Tris (MW 121.1) 60.5 g

NaCl 85 g

Distilled water 1 L

pH of the solution was adjusted to 7.6 and stored at 4oC.

0.1M Citrate Buffer

Citric acid 2.1 g

Distilled water 1 L

pH of the solution was adjusted to 6.0 and stored at 4oC.

Scott’s Tap Water

NaHCO3 24 mM

MgSO4 83 mM

CHAPTER 2

MATERIALS

56

The solution was stored at 4oC.

Gills Haemotoxylin

Ethylene glycol 25% v/v

Haematoxylin 6 g/L

NaIO4 2.8 mM

Aluminium sulphate.16H2O 80 mM

The ingredients were mixed overnight after which 6% v/v acetic acid was added. The

solution was then filtered and stored in the dark.

10% Phosphate Buffered Formalin Solution

Formaldehyde 10% v/v

The solution was made in PBS.

4% Paraformaldehyde

PBS was warmed in a microwave. Paraformaldehyde was added then to the PBS and

mixed vigorously to dissolve the powder.

DNase 1 buffer

Tris-HCl pH 8.0 40 mM

NaCl 10 mM

MgCl2 6 mM

CaCl2 10 mM

The solution was stored at 4oC.

DNase 1 (1 unit/mL)

DNase stock (10 unit/mL) 1 uL

DNase buffer 10 mL

Proteinase K buffer

Tris-HCl pH 8.0 100 mM

EDTA 50 mM

The solution was stored at 4oC.

CHAPTER 2

MATERIALS

57

2.3.3 FLOW CYTOMETRY

PBS/5% FCS/0.03%NaN3 (WASH BUFFER)

Heat inactivated FCS* 5 ml

NaN3 0.03 g

PBS 95 ml

*FCS was heat inactivated by keeping it in a water bath at 56oC for 2 hours. The wash

buffer was stored at 4oC.

Blocking buffer (20% normal goat serum)

Normal goat serum 2 ml

PBS 8 ml

PBS 90 mL

The solution was stored at 4oC.

2.3.4 CELL ISOLATION

RPMI/HEPES

HEPES 25 mM

RPMI 500 ml

The medium was stored at 4oC.

Percoll stock

Percoll 90%

10xPBS 10%

The solution was stored at 4oC.

70% Percoll

Percoll stock 70 ml

RPMI/HEPES 30 ml

The solution was stored at 4oC.

CHAPTER 2

MATERIALS

58

Ammonium Lysis Buffer

NH4Cl 82.9 g

KHCO3 10 g

EDTA 0.37

Distilled water 1 L

pH was adjusted to 7.2 and the solution was stored at 4oC.

2.3.5 ELISA REAGENTS

PBS/protease inhibitor

Protease inhibitor tablet supplied by Roche 1 tablet

PBS 10 mL

The solution was stored at 4oC and used within 2 weeks.

ELISA Wash buffer

Tween-20 0.5 mL

PBS 1 L

Stop solution

18M H3PO4 0.56 mL

H20 10 mL

CHAPTER 3

MATHODS

59

3.0 CHAPTER 3: METHODS

3.1 VIRUSES

3.1.1 VIRUS STRAINS

Flaviviruses that were used for in vitro and in vivo studies in this project were Kunjin

virus strain MRM16 (KUNV) (Doherty et al, 1963), Murray Valley encephalitis virus

strain OR2 (MVEV) (Leihne et al, 1976) and West Nile virus strain Sarafend (WNV)

(Scherret et al, 2001). These viruses were obtained from the existing departmental

stocks. They were originally derived from suckling mouse brain stocks and then

propagated twice in Vero cells. Glycosylation of the E protein may influence virulence

of a virus. Previously, it was shown that while KUNV MRM16 is a non-glycosylated

virus, WNV carries a glycosylated E protein (Scherret et al, 2001). However,

glycosylation of the E protein is also affected by the number of laboratory passage.

Thus, the actual glycosylation status of the viruses used in this project cannot be

provided as it was not examined and may vary between different laboratories.

Encephalomyocarditis virus (EMCV) was used in IFN type I bioassay.

3.1.2 PROPAGATION OF VIRUS STOCKS

For virus propagation, firstly, Vero cells were grown in 225 cm2 flasks (Nunclon

TM) in

30 mL of growth medium until cells were confluent. The spent medium was then

removed and cells were washed once with PBS. After removal of PBS, virus was added

to the cells at a multiplicity of infection (MOI) of 1 together with 10 mL of RPMI/2%

NCS. After 1 hour incubation at 37oC with occasional rocking, the inoculum was

removed and washed with PBS to remove unabsorbed virus. The cell culture flask was

added with fresh 30mL growth media containing 2% FCS and then incubated at 37oC.

Cytopathic effect was observed and when evident, the culture medium was removed and

centrifuged in Beckman Centrifuge (Model J-6B rotor JS-4.2) at 3000 rpm for 15 min at

4oC. The supernatant was removed and stored at –70

oC in 400 μL aliquots. Fifty percent

tissue culture infectivity dose (TCID50) bioassay was used to titrate the virus stocks and

titres were determined to be 7.9 log10 TCID50 units/100uL, 7.4 log10 TCID50

CHAPTER 3

MATHODS

60

units/100uL and 9.3 log10 TCID50 units/100uL for KUNV, MVEV and WNV,

respectively.

Methods for propagation of EMCV were similar except that the virus was grown in

murine L929 fibroblasts cells.

3.2 ANIMAL STUDIES

3.2.1 MOUSE STRAINS

Specific-pathogen free flavivirus susceptible C3H/HeJ (HeJ) and flavivirus resistant

C3H.M.domesticus-Flvr-like (DUB) inbred mouse strains were obtained from the

Animal Resource Centre (ARC), Murdoch, Western Australia. The young mice used

were from 3 to 4 weeks old while the adult mice were from 8 to 12 weeks old and they

were housed at the animal house in the Discipline of Microbiology and Immunology

under specific barrier maintained conditions with minimum disease risk. The cages were

changed once per week and infected mice were checked twice daily. Both male and

female mice were used in this project. All experimental work on mice was performed

according to the rules and practices prescribed by the UWA Animal Experimentation

and Ethics Committee.

3.2.2 VIRUS INOCULATION OF MICE

Different routes of inoculation were used in this project to infect mice. After infection,

mice were monitored and checked daily for signs of disease. The signs include

hunching, ruffling, hind leg paralysis and lethargy. Sick mice were sacrificed by

cervical dislocation before they experienced severe signs of diseases and became

moribund. Thus, any mortality or morbidity recorded in this project indicated deaths

that occurred before the onset of a moribund state.

3.2.2.1 Intracerebral inoculation

Virus stocks were diluted in sterile PBS. Virus was then administered intracerebrally

(i.c.) in 5 µL into penthrane-anaesthetised mice using a Hamilton repeating syringe

CHAPTER 3

MATHODS

61

fitted with a 26-gauge needle. The mice were inoculated under the eye, at the right side

of the head. Control mice were inoculated in a similar method with 5 μL PBS. Unless

otherwise stated, mice were infected with 100 times 50% lethal dose (100 LD50). The

LD50 is the dose that was required to kill 50% of adult HeJ mice and this dose varied

between KUNV and MVEV. Mice infected with KUNV received 1.74 x 105

infectious units (i.u.) while those infected with MVEV received 3.4 x 103 i.u.

3.2.2.2 Intraperitoneal inoculation

Adult and young HeJ and DUB mice were infected intraperitoneally (i.p.) with KUNV,

MVEV or WNV. Intraperitoneal injections were made into the caudal right abdominal

quadrant. Mice were restrained by hand and held with the body and head tilted

downward. The virus was diluted in sterile PBS and then injected using a 26G needle.

Some mice received treatment with LPS, thioglycollate, anti CD4+/CD8+ depleting

antibodies or SDS prior to or after virus inoculation (Section 3.3).

3.2.2.3 Intranasal inoculation

Intranasal inoculation (i.n.) of mice was performed by administrating 4 times the dose

was used for i.c. inoculation. The virus suspension was administered into the left and

right nares of penthrane-anaesthetised mice. A total volume of 20 l of virus containing

4 x 102 LD50 was given to each mouse.

3.3 INOCULATION OF REAGENTS/CELLS INTO MICE

3.3.1 THIOGLYCOLLATE

Thioglycollate is a sterile inflammatory agent and i.p. administration in mice results in

accumulation of macrophages in the peritoneal cavity. Six percent thioglycollate broth

was prepared in double distilled water, aliquoted into 10 mL and autoclaved. The

chemical was stored in dark at room temperature for at least 3 months prior to use in

animals. Adult mice were i.p. inoculated with 1mL of thioglycollate while young mice

CHAPTER 3

MATHODS

62

were i.p. inoculated with 200 uL. For both in vivo and in vitro studies, mice were

administered with thioglycollate 3 days prior to virus infection or cell isolation.

3.3.2 LIPOPOLYSACCHARIDE (LPS)

Lipopolysaccharide (LPS) was derived from Escherichi coli (E.coli) bacteria serotypes

0128:B12 (Sigma). The LPS was diluted in PBS at a concentration of 500 mg/mL and

mice received 100 uL by the i.p. route.

3.3.3 SODIUM DODECYL SULPHATE (SDS)

SDS was prepared in sterile double distilled water at a concentration of 2.4 μg/mL.

Mice were i.p. injected with 240 ng of SDS. For experiments involving administration

of the virus in conjunction with SDS, virus dilution was prepared in SDS solution.

Infectivity of the virus preparation was not affected by the presence of dilute SDS as

determined using the TCID50 bioassay.

3.3.4 CD4+ AND CD8 T+ CELLS DEPLETION

Mice were depleted of CD4 or/and CD8 T cells by i.p. inoculation of 150 μL anti-CD4

or 100 μL CD8 cell culture supernatant (equivalent to > 900ng antibodies) or both.

Unless otherwise stated, the depletion commenced 2 days before virus infection, and

then on day 2 and 4 p.i. in susceptible HeJ mice or on day 0, 2, 6 and 8 days p.i. in

resistant DUB mice. The monoclonal antibodies are cytotoxic and thus, would bind to

and cause apoptosis to the target cells.

To monitor the efficiency of T cell depletion, mice were given either 150 µL anti-CD4

or 100 µL anti-CD8 monoclonal antibodies on day 1 and day 3. On the fourth day,

spleens were harvested and splenocytes were isolated and labelled with anti-CD4+

(RL174, kindly donated by Dr J. Allen) and CD8+ (3.11 M, kindly donated by Dr J.

Allen) monoclonal antibodies that recognise different epitopes on T cells compared to

CHAPTER 3

MATHODS

63

the antibodies that were used to deplete T cells in mice (Lathbury et al, 1996).

Following this, flow cytometry analysis of the splenocytes was performed. The

depletion efficiency was determined to be 96% and 95% for CD4+ and CD8+ T cells,

respectively.

3.4.5 CLODRONATE

Clodronate (liposome encapsulated dichloromethylene-biphosphonate) was kindly

donated by Dr. Nico van Rooijen, Free University, Amsterdam, Netherlands and was

used to selectively deplete macrophages. HeJ and DUB mice were injected i.p. with

100 μL clodronate liposomes or liposomes only (negative control) before being infected

i.p. with WNV 4 days later. Intraperitoneal inoculation of clodronate depletes

macrophages in the peritoneal cavity, spleen, parathymic lymph nodes and liver (van

Rooijen and Sanders, 1994; Biewenga et al, 1995; Ciavarra et al, 1997). Efficiency of

macrophage depletion was assessed as discussed in Section 3.6.1.4.

3.4.6 MACROPHAGE TRANSFER EXPERIMENT

For adoptive transfer experiment, peritoneal macrophages were cultured under non-

adherent conditions for 5 days and later infected with WNV at MOI 10 (see Section

3.5.2.2 for isolation and culturing procedures). Two days after infection, the cultures

were centrifuged and supernatant was removed and stored at –80oC. The supernatant

was used to test the level of virus replication in peritoneal macrophages. Peritoneal cells

were washed with PBS and centrifuged twice before resuspended in PBS at

approximately 1 x 107 cells/100μL. Mice received 100uL of infected WNV and as

negative control, a separate group of mice were injected with uninfected macrophages.

3.4 ORGAN EXTRACTION

Mice were deeply anaesthetised by keeping them in an airtight jar filled with iso-

fluoropenthrane and they were sacrificed by cervical dislocation prior to organ

collection.

CHAPTER 3

MATHODS

64

3.4.1 BRAINS

Mice were sprayed with 70% alcohol and skin surrounding the head was removed. A

cut was made on top of the skull and then the skull was removed gently from the

cranium, ensuring that brain tissue was not damaged and all connecting muscle fibres

were removed. The brain was aseptically removed by sterile forceps into a collecting

tube. Brains were collected for viral titration, brain cell isolation, in situ cytokine

measurement or histology analysis. For virus titration, the collection tube contained

1mL cold RPMI medium with 2% NCS while for brain cells isolation, medium

comprised 1mL cold RPMI with 2% FCS and 25 mM HEPES. For cytokine

measurement and histology, collection tube was filled with 200 µL cold PBS/protease

inhibitor and 3 mL 10% buffered formalin saline, respectively. When brains were

extracted for cell isolation purposes, perfusion was performed prior to organ collection

to remove contaminating erythrocytes and non-brain infiltrating cells. In these instances,

the chest cavities of anaesthetised mice were opened. A butterfly clip 26G needle was

inserted into the lower left ventricle of the heart and mice were perfused with 10 to

20 mL of cold PBS. The brains were extracted and kept on ice until they were

homogenised. For histology analysis, brains were kept in 10% buffered formalin saline

at room temperature for 48 hour before being processed at the Department of Pathology,

UWA.

3.4.2 PERIPHERAL ORGANS

Mice were pinned to a corkboard with the ventral side facing up. Seventy percent

alcohol was sprayed on the body and skin was cut from the abdomen to the chest. The

skin was removed, exposing the peritoneal wall. A cut was then made with sterile

scissor to open the peritoneum. Organs including spleen, liver, kidney and pancreas

were removed gently and aseptically into small tubes containing cold RPMI with 2%

NCS or FCS. For virus titration, organs were homogenised into 10% extracts using

sterilised glass tissue grinders. For detection of macrophages in spleens, these organs

were embedded in tissue-tek and stored at –80 oC until ready for cryosectioning.

CHAPTER 3

MATHODS

65

3.5 CELL ISOLATION

3.5.1 BRAIN MONONUCLEAR CELLS

Brains were transferred from collection tubes to cold sterile glass tissue homogenisers.

1mL RPMI with 25 mM HEPES was added and brain tissue was ground to a smooth

consistency. The homogenate was then transferred into a pre-chilled 15 ml centrifuge

tube. Two brains were used for each 15 mL centrifuge tube. The volume in the

centrifuge tube was adjusted to 7 mL by adding extra medium. Then, 3 mL of stock

Percoll was added and the contents mixed by inversion. This resulted in a 30% Percoll

solution. One mL of 70% Percoll was added slowly to the bottom of the centrifuge tube,

using a sterile 1 mL glass pipette so that the 30% Percoll solution at the top of the tube

was not disturbed, resulting in a sharp interphase. The tube was centrifuged at 2000 g

for 20 min at 20oC without applying a brake (Beckman J6-MI centrifuge). Following

centrifugation, the top phase which contained myelin was removed using a 25 mL glass

pipette. Mononuclear cells were in the interphase and lay above the 70% Percoll. After

removing most of the top phase (30% Percoll solution), the interphase was transferred

gently to a fresh 15 mL tube without disturbing (if any) red blood cells that formed a

pellet at the bottom of the tube. Cells were washed in 10 to 12 mL of medium after

being transferred into a fresh tube and centrifuged again at 4oC for 12 min. Supernatant

was removed and cell pellet were resuspended in a final volume of 300 uL. When cells

were used for flow cytometry, they were resuspended in PBS containing 5% heat

inactivated FCS and 0.03% NaN3. For studies involving culturing the cells, RPMI with

10% or 5% FCS was used, respectively.

3.5.2 SPLENOCYTES

Spleens were cut into smaller pieces and transferred to a tissue sieve, which was kept on

top of a cold 50 mL centrifuge tube. Using 10 mL plunger, the spleens were gently

squashed to remove the splenocytes. Following this, RPMI/2% NCS was added to wash

the sieve. Cells were collected at the bottom of 50 mL centrifuge tube. The sieve was

washed 3 times with 2 mL medium. The tube was centrifuged at 3000 rpm at 4oC for

12 min. After centrifugation, supernatant was removed and cell pellet was resuspended

CHAPTER 3

MATHODS

66

in 2 mL lysis buffer. Lysis buffer was used to remove contaminating red blood cells.

The cells were incubated at room temperature for 2-3 min after which 10 mL medium

was added to stop the cell lysis. The cells were centrifuged twice. Cells were finally

resuspended in 1 mL wash buffer prior to flow cytometry analysis.

3.5.3 PERITONEAL MACROPHAGES

3.5.3.1 In vitro experiments

Three days after administrating 6% thioglycollate, mice were culled by cervical

dislocation and pinned to a corkboard. Skin was sprayed with 70% alcohol and a cut

was made at the abdomen to expose the peritoneal wall. 5 mL of cold sterile PBS was

carefully injected into the peritoneal cavity using a 26G needle. The peritoneal wall was

then massaged gently to help dislodge the peritoneal cells. The cells were later aspirated

using the same needle slowly and carefully, so that internal organs were not punctured

or damaged by the needle. The process was repeated and the peritoneal fluid was

transferred into cold 50 mL centrifuge tubes. Peritoneal cells were centrifuged

(Beckman J6-MI centrifuge) at 1500 rpm for 5 min at 4oC and cell pellet was

resuspended in cold PBS and centrifuged again. Cells were resuspended in 3 to 5 mL of

RPMI with 10% FCS. Trypan Blue exclusion dye was then used to determine cell

number and viability.

Peritoneal macrophages were usually pooled from several mice and cells were grown

either under adherent or non-adherent conditions. Adherent peritoneal cells were used to

study viral replication and MHC molecules up-regulation following KUNV, MVEV and

WNV infections. To prepare this, cells were seeded into 6 well culture tissue plates at a

density of 1 x 106 cells/well. The peritoneal cells were incubated overnight in a total

volume of 3 mL medium/well at 37oC and in a 5% CO2 humidified atmosphere. The

next day, non-adherent cells were removed by washing the wells vigorously, twice with

PBS. Peritoneal macrophages adhered to the tissue culture flask and these cells were

further incubated in fresh 2 mL RPMI with 10% FCS and later used for various

analyses. For a non-adherent growth condition of peritoneal cells, they were grown in

teflon pots at 1 x 106 cells/mL. Every two days in culture, cells and spent media were

CHAPTER 3

MATHODS

67

transferred into 50 mL centrifuge tubes and centrifuged for 5 min at 2000 rpm. Cells

were washed twice in 10 mL PBS and they were finally resuspended in fresh RPMI

containing 10% FCS and cultured in the teflon pots. Peritoneal cells grown in either

adherent or non-adherent conditions were cultured for a maximum period of 5 days after

isolation prior to virus infection. This allowed the removal of cytokines that may be

produced by macrophages during thioglycollate treatment or isolation which could

affect virus infection of the cells.

3.5.3.2 In vivo experiments

For cell transfer studies, peritoneal macrophages were isolated as mentioned above.

However, following collection of the cells, peritoneal macrophages were centrifuged

and washed twice with PBS. The cell numbers and viability were determined and the

peritoneal cells were resuspended in 10% RPMI at a density of 1 x 106 cells per mL.

Five to six mL of cell suspensions were transferred to teflon pots to maintain the culture

in a non-adherent condition as described above. The media were regularly changed

every two days by centrifuging cells for 5 min at 2000 rpm and washing the cells twice

with PBS. Following this, fresh media was added to the cells. Peritoneal macrophages

were cultured for 5 days prior to virus infection.

CHAPTER 3

MATHODS

68

3.6 HISTOLOGICAL PREPARATION AND IMMUNOHISTOCHEMISTRY OF

ORGANS

3.6.1 BRAIN

3.6.1.1 Paraffin embedding of the brain

Brains were removed aseptically and kept in 10% buffered formalin saline, as

previously described (section 3.4.1) and left at room temperature for 48 hours before

they were taken to the Department of Pathology (UWA) for further processing into

paraffin blocks. Paraffin embedded brain tissue was cut in 10μm saggital and horizontal

orientations and placed onto 3-aminopropyltriethoxysilane pre-coated microscope slides

(silanated slides). The slides were kept at room temperature prior to staining.

3.6.1.2 Hematoxylin and eosin (HE) staining

Staining of brain tissue sections with HE was performed with assistance from staff in

the Department of Pathology. Briefly, the slides were deparaffinised by washing

consecutively in xylene, 100%, 95% and 70% ethanol, and water. Gill’s hematoxylin

was then added for 3 min after which the slides were washed sequentially in water,

Scott’s Tap water and ethanol. Eosin was added for 1 min and the slides were washed in

ethanol, ethanol: xylene and xylene. The slides were mounted onto permanent mounting

medium and covered with cover slips. Using these slides, the study of brain architecture

and inflammatory responses was undertaken under the guidance of Professor John

Papadimitriou at the Department of Pathology, UWA. Photographs were taken using a

light microscope with an attached digital camera (Leica Digital Camera and Viewer).

CHAPTER 3

MATHODS

69

3.6.1.3 Activated brain microglia/macrophages labeling

Labeling of activated microglial/macrophage cells in the brain was performed by

utilising tomato lectin (Lycopersicum Esculentum). Brain tissue sections placed on

silanated slides were consecutively washed with 100% xylene, 1:1 xylene and alcohol,

100% alcohol, 70% alcohol, 50% alcohol and water. The slides were then placed in

citrate buffer and heated in a microwave for 2 x 4 min to retrieve microglia antigen.

Following this, the brain sections were washed in TRIS Buffered Saline (TBS) and then

incubated in 0.3% hydrogen peroxidase for 30 min. Further treatment involved

incubating the brain sections with biotinylated tomato lectin for 2 hours. Upon washing

with TBS, streptavidin conjugated with hydrogen peroxidase was applied onto each

slide and incubated for 10 min. Following incubation, DAB solution was added to the

slides for 3 to 5 min before being washed with distilled water to stop the reaction. Slides

were counterstained with haematoxylin (kindly provided by staff at the Department of

Pathology, UWA) for 30 sec, washed in distilled water, and then in Scott’s tap water for

30 sec before being washed again in distilled water. Slides were later washed

sequentially for 3 min in 50% alcohol, 70% alcohol, 100% alcohol, ethanol:xylene and

100% xylene, and finally mounted using DEPEX mounting solution and left overnight

to dry.

3.6.1.4 Detection of macrophages in spleens

To confirm that clodronate treatment depletes macrophages in mice (Section 3.3.5),

mice were administered with 100 μL clodronate and two days later, spleens were

harvested from these mice and embedded in tissue-tek prior to storage at –80 oC.

3.6.1.4.1 Cryosectioning of spleen

Blocks of embedded tissues were placed in the cryostat (Leica CM 1900) at –20oC for

15 min to allow the samples to equilibrate to the temperature of the cryostat. Spleens

were placed on the chuck and mounted using tissue-tek. Ten μm sections were prepared

from these organs, placed onto silanated slides and kept at –20oC.

CHAPTER 3

MATHODS

70

3.6.1.4.2 Detection of macrophages

Detection of macrophages was performed using a rat raised monoclonal antibody

specific for F4/80 cell surface antigen which is expressed by macrophages (Austyn and

Gordon, 1981). The frozen tissue sections from control and macrophage-depleted mice

were air-dried prior to being fixed in cold methanol for 10 min. Following this, tissue

sections were washed 3 times for 2 min and then incubated in blocking solution for 30

min. Spleen tissue sections were then removed from the blocking solution and applied

with the primary antibodies, F4/80 (neat culture supernatant). Tissue sections from

clodronate-treated mice were not only used to assess the efficiency of macrophage

depletion, but were also employed as controls for background reaction of F4/80

antibodies. To ensure that secondary antibody bind specifically to the primary antibody,

a few tissue sections from control mice were applied with Tris saline buffer (TSB)

rather than F4/80 antibodies (negative controls). Following 2 hours incubation, all slides

were washed with TSB 5 times (5 min per wash) and then incubated with biotinylated

goat anti-rat IgG (1/200 dilution). After 45 min incubation, slides were washed and

streptavidin-horseradish-peroxidase was added to the slides for 10 min. Slides were

washed again and later incubated with DAB for 5 min. To stop the reaction, slides were

immersed in distilled water for 10 min. Hematoxylin was then used to counterstain the

tissue sections. Following this, the slides were washed for 3-5 min in 70%, 85%, and

100% ethanol, ethanol:xylene and lastly in 100% xylene. The slides were mounted

using DEPEX mounting solution and left overnight to dry.

3.6.1.5 Apoptosis Detection

Apoptotic cells were detected using the DeadEnd Colorimetric TUNEL System

(Promega). The brain tissue sections on pre-coated slides were first deparaffinised in

xylene for 5 min. The slides were washed through graded ethanol (100%, 95%, 85%,

70% and 50%), 0.85% NaCl and PBS for 5 min in each wash. To fix the tissue, the

tissue sections were incubated in 4% paraformaldehyde for 15 min and then washed

with PBS. Proteinase K was added for 10 min to permeabilise the tissues. Slides were

again washed in 4% paraformaldehyde before addition of 100 μL Equilibration buffer

(supplied). Both positive and negative controls were also included in this assay. Positive

control was prepared by treating brains tissue sections with DNase I to induce DNA

fragmentation after washing with the slides with paraformaldehyde. Brain tissue

CHAPTER 3

MATHODS

71

sections from uninfected mice were used as negative control. After 10 min incubation,

biotinylated nucleotide mix (supplied) was added and the slides were incubated at 37oC

for one hour. The reaction was stopped by immersing slides in 2 x SSC for 15 min and

subsequent washing in PBS. The slides were then added with hydrogen peroxide (0.3%)

to block the endogenous peroxidase. After that, 100 μL of streptavidin conjugated with

hydrogen peroxidase (HRP) was dispensed onto each slides and incubated for 30 min.

Following incubation, slides were washed with PBS and DAB components were added.

Dark brown colour developed in 13 min in tissues containing apoptotic cells. To

counterstain the slides, Gill’s Hematoxylin (kindly provided by staff at the Department

of Pathology) was used.

3.7 CELL STUDIES

All cell lines used in this research were obtained from departmental stocks.

3.7.1 AFRICAN GREEN MONKEY KIDNEY CELLS (VERO CELLS) AND L292

MOUSE FIBROBLASTS

Vero and L292 cells were grown in RPMI 1640 media containing 10% FCS in 225 cm2

tissue culture flasks (NunclonTM

) until confluent. To subculture the cells, the spent

medium was removed and cells were washed with 10 mL PBS. Following that, 10 mL

PBS/trypsin/EDTA was added, incubated for 1 minute and then removed, leaving 1 mL

in the flask. The flasks were incubated further for 4 min at 37oC. Ten mL medium was

added and the cells were resuspended gently to break up cell clumps. One mL of cell

suspension was used to seed the flask. Fresh growth media (30 mL) was added to the

flask and cells were incubated at 37oC.

3.7.2 HYBRIDOMA YTS 191 AND 169 CELL LINES

3.7.2.1 Cell culture

Hybridoma cells lines YTS 191 and YTS 169 (American Type Tissue culture (ATTC),

kindly provided by Dr. Anthony Scalzo, Lions Eyes Institute) (Lathbury et al, 1996;

Cobbold et al, 1984) were grown in RPMI 1640 containing 10% FCS/2mM

CHAPTER 3

MATHODS

72

glu/0.05mM 2ME/1mM sodium pyruvate. Cells were subcultured every 3 days. Since

these cells are semi adherent, to subculture, cells were dislodged from the flask by

washing the flask several times with the spent media. If washing alone could not

dislodge the cells, 3 mL PBS containing trypsin then were added and flask was

incubated for 1 to 2 min. Spent media containing cells were then transferred to 50 ml

centrifuge tubes and centrifuged at 2400 rpm for 5 min. The supernatant was removed,

cells were resuspended in 10 ml media and 1 ml was returned to the flask. An additional

35 ml of medium was added and cells were incubated at 37oC with 5% CO2.

3.7.2.2 Production of anti CD4+ and anti CD8+ antibodies

For production of anti CD4 and anti CD8 antibodies, YTS 191 (produces anti-CD4,

IgG2b) and YTS169 (produces anti-CD8, IgG2b) hybridoma cell lines were cultured

in 75 mL media for 4 days until confluent. On day 4, spent media were removed and

centrifuged in 50 ml centrifuge tube. Cells were resuspended in 50 ml

RPMI/glu/2ME/sodium pyruvate without FCS for the next 4 days. The absence of FCS

caused cells to eventually die, resulting in the secretion of antibodies into culture

supernatant. On day 4, when signs of cell deaths were apparent, spent media were

collected and stored at 4oC prior to further processing.

3.7.2.3 Ammonium sulfate precipitation

Ammonium sulfate precipitation was used to purify and concentrate the antibodies. In

solution, proteins form hydrogen bonds with water through their exposed polar and

ionic groups. When high concentrations of small, highly charged ions such as

ammonium or sulfate are added, these groups compete with the proteins for binding

with water. This removes water from the proteins, eventually resulting in precipitation.

Total volume of tissue culture supernatant (spent media containing antibodies) was

determined prior to centrifugation at 3000 g for 30 min. The supernatant was then

transferred to a 2 L beaker which was kept on a magnetic stirrer. While the antibody

solution was being stirred gently, saturated ammonium sulphate was added drop wise to

bring the final concentration of this solution to 50%. The solution then turned cloudy,

indicating that antibodies had precipitated. The beaker was then transferred to a cold

room (4oC) and kept overnight. The following day, the precipitate was centrifuged at

CHAPTER 3

MATHODS

73

3000 g for 30 min. The supernatant was discarded and the pellet was suspended with

PBS (0.1 x the starting culture supernatant volume). The antibody solution was then

transferred to sterile dialysis tubing measuring approximately 20 cm. The dialysis

tubing was kept in a beaker, containing 2L PBS and stirred overnight at 4oC. PBS was

changed 3 times in 2 days. The antibody solution was then removed from the tubing and

centrifuged. Finally the antibody was filter sterilised through a 0.22 μm syringe filter,

aliquoted into 1.5ml eppendorf tubes and kept at –20oC. Protein concentration was

determined using a UV spectrophotometer at 280 nm with the assumption that optical

density of 1.35 was equal to 1 mg/ml protein. The concentration was found to be

600 µg/ml and 926 µg/ml for anti CD4+ and anti CD8+ culture supernatant,

respectively.

3.7. 3 MOUSE PRIMARY CELL CULTURES

Primary mouse macrophages were cultured in growth medium (RPMI) containing 10%

FCS and 2 mM glutamine. For dendritic cells (DCs) (kindly provided by Ms Andrea

Kong), cells were grown in growth medium (DMEM) containing 10% GM-CSF

(Kindly provided by Ms Andrea Kong). Cells were either cultured in 6 well culture

plates (at 1x106 cells/well) or in teflon pots (at 1x10

6 cells/mL).

3.7.4 VIRUS INFECTION OF CELLS

Primary cell lines were grown in growth media in 6 well culture tissue plates (adherent)

(1x106 cells/well) or teflon pots (non-adherent) (1x10

6 cells/mL), depending on the

experiment. To infect cells with virus, cells were first washed with sterile PBS gently.

Cells were then infected with virus at MOI 10, diluted in mL RPMI with 2% FCS for 1

hour (1 x 107 i.u./mL). This value (1 x 10

7 i.u./mL) is equivalent to 10

6.2/100μL TCID50

units. The plates or the pots were incubated at 37oC in 5% CO2 with occasional rocking

to facilitate virus binding to the cells. Following one hour incubation, medium

containing free unattached virus was removed and cells were washed with PBS twice.

Fresh 2mL medium with 2% FCS or NCS was then added into each well of the 6 well

plates. After a gentle rocking, 0.5mL to 1mL aliquots were immediately taken from the

wells and replaced with similar volume of fresh media. These samples were used to

measure virus titres at time 0. At various time points after infection, aliquots of

CHAPTER 3

MATHODS

74

supernatant from 6 well plates were removed, stored at –80oC and later used for virus

titration. For cells cultured in teflon pots, 1mL of new medium was added per 1 x 106

cells. Virus titration was performed by TCID50 bioassay.

3.7.5 PREPARING CELL STOCKS

To prepare stocks of the cells used in this project, cells were grown in appropriate

growth medium until confluent. Cells were removed from the tissue culture flask and

centrifuged at 4oC at 2000 rpm for 15 mins. Following centrifugation, supernatant was

discarded and cells were suspended in 9 mL cold FCS. Nine mL of 10% dimethyl

sulphoxide (DMSO) in FCS was then added slowly to the cells, avoiding any bubbles.

Cells were aliquoted into 1.8 mL cryotubes and kept at –70oC overnight. The following

day, cryotubes were transferred into liquid nitrogen.

3.7.6 VIRUS TITRATION

3.7.6.1 Preparation of 10% brain homogenates

Brain was removed aseptically and kept in cold RPMI containing 2% NCS. The brain

was weighed and 10% brain homogenates were prepared aseptically using cold glass

homogenisers in fresh medium. The homogenates were then centrifuged at 3000 g, for

15 min at 4oC (Beckman Centrifuge, Model J-6B rotor JS-4.2). Supernatant containing

the virus was removed and stored in 500 μL aliquots at –70oC.

3.7.6.2 Tissue culture infectivity dose 50% (TCID50)

The titres of virus present in harvested organs or culture supernatant was determined by

TCID50 assay (where the TCID50 is the highest dilution of virus that causes cytopathic

effect in 50% of cell cultures). In this assay, Vero cells were used as indicator cell lines.

Wells of 96 well plates (MICROTESTTM

Tissue Culture) were seeded with 100 μL of

Vero cell suspension, containing approximately 2 x 105 cells/mL and incubated at 37

oC

in 5% CO2 until 90% confluent (1-2 days). Virus samples from 10% homogenates or

culture supernatant were diluted from 10-1

to 10-9

in RPMI containing 2% NCS in a 25-

CHAPTER 3

MATHODS

75

well sterilin tray. The growth medium was removed from the 96-well plates and 100 μL

of each virus dilution was added to 10 wells using a multi-channel. Two wells were

included as cell control, into which only media was added. The cells were incubated at

37oC for 7 days and checked occasionally for CPE. On day 7, the presence of CPE was

determined using an inverted microscope and confirmed by adding 100 μL of methylene

blue into the wells. The plates were left overnight, and then washed and rinsed with tap

water. The number of wells that showed CPE was recorded and the TCID50 for each

sample was determined using an in-house computer programme.

The sensitivity of this assay in detecting accurate virus titres is at 2.0 log10 TCID50

units/100μL. This value corresponds to 0.7 x 102 i.u./100µL. As virus is also present

below 2.0 log10 TCID50 units, the amount of virus was expressed by 2 arbitrary values;

1.0 log10 TCID50 units for virus detected between 0.7 x 102 i.u./100µL and 0.35 x 10

2

i.u./100µL, and 0.5 log10 TCID50 for virus detected between 0.35 x 102 i.u./100µL and

0.1 x 102 i.u./100µL. This assay would not detect any infectious virus below 0.1 x 10

2

i.u./100µL.

3.8 CYTOKINE STUDIES

3.8.1 IFN TYPE I BIOASSAY

This assay tests for the presence of IFN type I in the samples by utilising the antiviral

properties of this cytokine following infection of indicator cell line L929 mouse

fibroblasts with EMCV.

3.8.1.1 Preparation of L929 monolayers

L929 cells were grown until confluent, trypsinised and resuspended in RPMI containing

10% NCS. One hundred μL of resuspended cells were seeded onto wells of 96-well

plates and incubated at 37oC until confluent (1-2 days).

CHAPTER 3

MATHODS

76

3.8.1.2 Acid treatment of samples

Since IFN assay is based on the biological (antiviral) property of IFN type I, acid

treatment was performed in order to destroy other constituents of the tissues that may

have antiviral properties or infectious virus that could infect and kill L929 cells and

eventually would lead to inaccurate results. Five hundred microlitres of samples were

acidified to pH 2 by adding 50-60 μL HCl (0.5M). At pH 2, most proteins are denatured

and precipitated while virus is inactivated. IFN type 1 however is not affected and

exerts its full biological activity when the pH is re-adjusted to 7. Thus, following one-

hour incubation, samples were neutralised to pH 7 by adding 40-50 μL NaOH (0.5 M)

and then centrifuged at 3500 g for 15 mins. Supernatants were collected and stored at

–70oC.

3.8.1.3 IFN type I bioassay

Brain homogenates and IFN standard were diluted 1:6 in RPMI containing 2% NCS,

and 250 μL of this dilution was transferred in duplicate to the first rows of the first

96-well plates (which did not contain any cells). One hundred and twenty five μL of

RPMI containing 2% NCS was dispensed to the rest of the wells and serial two-fold

dilutions were performed across the plates. The spent medium in the second 96-well

plates that contained confluent monolayer L929 cells (as prepared in 3.8.1.1) was

discarded and then plates were washed with sterile PBS. PBS was removed and 100 μL

of prepared diluted samples and standard IFN was transferred from the first 96-well

plates into the corresponding wells of plates containing L929 cells. Positive and

negative controls were also included in each plate at this stage. The positive controls

contained virus only, while negative control contained medium only (6 wells per

control). The plates were then incubated at 37oC in 5% CO2 for 20 hours.

Following incubation, the samples were discarded and the 96-well plates were washed

with sterile PBS. Stock EMCV was diluted to 10-6

in RPMI with no serum and 100 μL

was added to all wells excluding the negative control wells. Only RPMI was added into

these negative control wells. Plates were then incubated again until CPE could be

observed in positive control cells (> 20 hours). The plates were stained with methylene

blue-formaldehyde, left overnight, washed and wells with 50% CPE were determined.

The 50% CPE in the samples were determined by comparing test wells with the 50%

CHAPTER 3

MATHODS

77

CPE observed in IFN standard with known IFN type I concentrations and values were

expressed at international units/mL (I.U./mL). The sensitivity of this assay is

2.5 I.U./mL.

3.8.2 CYTOKINE ELISA

For detection of TNF, IFN, IL-2, IL-4 and IL-10, a commercial ELISA was used

(eBioscience). High protein binding capacity 96 well flat bottom plates (such as

Corning Costar 9018) were coated with 100 μL per well of capture antibody, which had

been diluted in coating buffer as recommended by the supplier. The plates were sealed

and kept overnight at 4C. The next day, the contents were removed and plates were

washed 3 times with 300 μL per well with wash buffer. Plates were then blotted on

absorbent paper to remove any residual buffer. Following this, plates were blocked with

100μL per well of assay diluent for one hour at room temperature. Plates were then

washed with wash buffer as before. 100 μL per well of standard was added to the

appropriate wells and 2-fold serial dilutions were performed to construct the standard

curve. 100 L of test samples were added to the wells and incubated at room

temperature for 2 hours. Plates were washed 5 times and 100 L detection antibody

(conjugated with biotin )was added to the wells and further incubated for 1 hour. Then,

plates were washed 5 times and Avidin-Horseradish peroxidase was added (100

L/well). The plates were left at room temperature for 30 min before being washed 7

times. During this step, wells were soaked with wash buffer for 1-2 min each wash. To

develop colour, 100 L substrate solution (tetramethylbenzidine, TMB) was distributed

into each well and left for 15 min. Fifty L stop solution (1M H3PO4) was added to stop

the reaction and absorbance was measured at 450 nm using Bio-Rad Model 3550

Microplate Reader. The amount of cytokines present in the samples was determined

using the standard curve.

3.9 FLOW CYTOMETRY

Following isolation of mononuclear cells and splenocytes, numbers of cells were

determined using trypan blue dye and the cells were resuspended at 1 x 107 cells/mL in

CHAPTER 3

MATHODS

78

wash buffer. 100 μL was added to the wells of 96-well round bottom plate. Plates were

centrifuged at 200 g for 5 mins at 4C and the supernatant was removed. Cells were

washed twice and incubated for 30 min in blocking buffer (PBS/20% normal horse

serum) on ice. After washing the cells twice with wash buffer, 100 µL of rat anti CD4,

CD8, CD19, CD45, CD11b, CD11c, MHC class I or MHC class II antibodies that were

diluted 1/500 in wash buffer were added to the cells and incubated on ice in a dark for

60 min. These antibodies were directly conjugated with fluorescein dye (either PE or

FITC). Following this, cells were washed twice and then finally resuspended in 500 uL

wash buffer. Unstained cells were included as a negative control. Cells were kept on ice

in the dark until further analysis by flow cytometry.

For IFN intracellular staining, cells were incubated in RPMI/ 5%FCS/ 2mM glutamine/

2 μg per mL Brefeldin A for 4 hours prior to labelling the cells with monoclonal

antibodies. Brefeldin A interferes with protein transport from the endoplasmic reticulum

to the Golgi apparatus, leading to the accumulation of protein in the endoplasmic

reticulum. This allows detection of proteins of interest following intracellular staining.

After incubating cells with anti-CD4+, CD8+ or CD11b+ monoclonal antibodies for 30

min, cells were washed twice with wash buffer. Cells were then fixed in 4%

paraformaldehyde for 30 min on ice in the dark. Following this, cells were washed once

in wash buffer and 100 μL 0.1% saponin in PBS was added for 30 min to permeabilise

the cells. After two washes with PBS, cells were incubated with anti IFN antibody for

60 min. Cells labelled with isotype antibody were included as negative control.

For flow cytometry analysis, cells were analysed in the Biomedical Imaging and

Analysis Facility, UWA, using a Becton-Dickinson FACSCalibur. The cells were gated

as defined by forward and side scatter to ensure that 10,000 cells were being recorded,

and analysis was performed using the CELLQUEST program.

CHAPTER 3

MATHODS

79

3.10 STATISTICAL ANALYSIS

Student T-test was used in this project for statistical analysis of brain viral titres, ATD

and cytokine levels (Excel program). ANOVA was used when analysis involved more

than two groups. For analysis of mortality rate of mice, Kaplan Meier was employed

(SPSS program).

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

80

4.0 CHAPTER 4: STUDY ON KUNV, MVEV AND WNV

VIRULENCE IN SUSCEPTIBLE AND CONGENIC

RESISTANT MICE

4.1 INTRODUCTION

Numerous data have demonstrated that the outcome of infections is contributed by

many different host and virus factors (Brinton and Perelygin, 2003). Virulence of a

virus is the most prominent viral feature that in concert with a viral dose and route of

inoculation, could dictate the severity of disease. The virus virulence is characterised by

the ability of virus to invade the CNS (neuroinvasiveness), and by the ability of virus to

establish lethal infection in the brain (neurovirulence).

The neurovirulence of several flaviviruses belonging to the JE complex serogroup has

been studied earlier in this laboratory in flavivirus susceptible HeJ and congenic

resistant MOLD, RV and DUB mice. Resistant MOLD and DUB mice, developed in

this laboratory, carry novel resistance alleles Flvr-like and Flv

mr, respectively, which

confer different degrees of resistance against flaviviruses when compared to resistant

RV mice (Urosevic et al, 1999). Data from this and other laboratories indicated that the

same flavivirus may produce different neurovirulence patterns in susceptible and

resistant mice (Urosevic et al, 1999; Sabin, 1952a). For example, i.c. challenge of 17D

YFV resulted in 100% mortality in HeJ mice but did not affect the survival of resistant

mice (Urosevic et al, 1999; Sabin, 1952a). In contrast, i.c. challenge with MVEV strain

1/51 induced less than 50% death in both susceptible and resistant mice (Shueb et al,

2005).

The aim of the study described in this chapter was to extend our knowledge and to shed

further light onto the neurovirulence and neuroinvasive properties of three flaviviruses,

WNV (Sarafend), MVEV (OR2) and KUNV (MRM16) in two mouse strains expressing

different levels of susceptibility/resistance to flaviviruses; the flavivirus susceptible HeJ

and flavivirus resistant DUB mice. Although KUNV, MVEV and WNV have been

extensively used previously in this laboratory, their virulence in resistant and

susceptible mice has never been completely characterised. Thus, to assess their

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

81

neuroinvasiveness and neurovirulence, these three flaviviruses were delivered by the i.p.

and i.c. routes to susceptible HeJ and resistant DUB mice. Additionally, a small study

involving intranasal (i.n.) infection was also performed in parallel to examine virus

virulence in the absence of mechanical (needle) injury to the CNS.

In addition, the second aim of the study was to investigate the effect of breaching BBB

or alteration of function/population of host innate and adaptive immune cells on the

neuroinvasiveness of the three flaviviruses. To achieve this, two chemicals, SDS and

LPS, which are known to modulate the BBB were assessed for their effect on virus

neuroinvasiveness. Host early defence/inflammatory cells were also manipulated using

treatments with thioglycollate, clodronate and anti-T cells antibodies to examine

whether the absence/presence of certain cell types would facilitate flavivirus invasion to

the brain and subsequently induce a more severe disease in mice.

4.2 RESULTS

4.2.1 VIRUS NEUROVURULENCE STUDIES

This study was undertaken to compare the neurovirulence of WNV, KUNV and MVEV

in flavivirus HeJ susceptible and resistant DUB mice following i.c. challenge. This

work was aimed at determining whether a difference in the neurovirulence displayed by

these viruses in susceptible mice would also be displayed in flavivirus resistant DUB

mice. The viruses used in this study were propagated once in suckling mouse brain and

twice in Vero cells. TCID50 bioassay was performed to determine the titres of the virus

stocks. Initial stocks of WNV, KUNV and MVEV were determined to have titres of

109.2

/100 µL, 107.9

/100 µL and 107.4

/100 µL TCID50 units respectively.

4.2.1.1 Analysis of neurovirulence of WNV, KUNV and MVEV in susceptible mice

In this study, virus stocks were serially diluted in PBS from 10-1

and up to 10-6

.

Following this, 5 µL of each virus dilution were inoculated by the i.c. route to

susceptible HeJ and resistant DUB mice. Groups consisting of 8-20 mice of both sexes

aged between 12 to 15 weeks were used per each virus dilution, and the mice were

monitored for signs of disease for up to 30 days p.i. Signs of illness exhibited by sick

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

82

mice included ruffled fur, hunched back, body weight reduction and hind leg paralysis.

Sick animals were culled and deaths were recorded before they reached moribund stage,

in accordance with the rules and practices prescribed by the UWA Animal

Experimentation and Ethics Committee. As negative controls, two groups of 5-8 mice

were inoculated i.c. with either PBS or UV-treated viruses. These mice did not exhibit

any signs of disease when monitored for 30 days indicating that the injection alone or

dead virus could not trigger fatal encephalitis in mice.

As shown in Table 4.1.A, the outcome of infection, which is defined by the mortality

rates and average time to death (ATD) of infected animals, was affected by the dose and

type of infecting viruses. High doses of KUNV induced 100% mortality in HeJ mice, as

seen in animals i.c. infected with 3.0 x 105 , 3.0 x 10

4 or 3.0 x 10

3 infectious units (i.u.)

of KUNV. In contrast, mice were not affected when infected with weaker doses; 3.0 x

102 i.u. of KUNV or lower (virus stock dilution 10

-3 and higher). Similarly, all HeJ mice

had fatal encephalitis following i.c. challenge with 8.0 x 105 , 8.0 x 10

4 or 8.0 x 10

3 i.u.

of WNV (10-2

to 10-4

virus stock dilutions) while they displayed complete survival with

lower amounts of WNV (8.0 x 102 i.u. and 8.0 x 10 i.u. of WNV). Interestingly, lower

doses of MVEV compared with KUNV and WNV were required to cause similar 100%

lethal infection in susceptible HeJ mice (1.0 x 103 i.u. and 1.0 x 10

2 i.u. of MVEV),

suggesting that these flaviviruses, although closely related, exhibit disparate

neurovirulence in susceptible HeJ mice.

To investigate whether different strains of susceptible mice would be uniformly

vulnerable to flavivirus infection, parallel i.c. infection was also carried in three

flavivirus susceptible mouse strains; HeJ, C57BL/6 and BALB/c mice. However, for

this study, only two dilutions of KUNV were used. As shown in Table 4.1.B, C57BL/6

mice displayed a slight resistance to i.c. KUNV challenge as suggested by the lowest

mortality recorded compared with the other mouse strains, HeJ and BALB/c that had

similarly high mortality rates. However, the difference in mortality rates and ATD

observed in these animals was not significant (Student t test, p > 0.05)

.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

83

Table 4.1.A Mortality and LD50 studies following intracerebral infection with

serially diluted viruses in flavivirus susceptible HeJ mice.

Virus

Virus

stock

dilutions

Amount

(i.u.)a

Mortality/

tested ATD

b

LD50

(virus

dilutions)

LD50

(i.u.)

KUNV

10-1

3.0 x 105 20/20 5.0 ± 0

10-3.5

1.7 x 103

10-2

3.0 x 104

10/10

5.0 ± 0

10-3

3.0 x 103 10/10 5.0 ± 0

10-4

3.0 x 102 0/10 -

10-5

3.0 x 101 0/10 -

MVEV

10-3

1.0 x 103 10/10 6.0 ± 0

10-4.62

3.4 x 101

10-4

1.0 x 102 10/10

7.4 ±

1.4

10-5

1.0 x 101 2/10 8.0 ± 0

10-6

1.0 x 100 2/10 11.0 ± 0

WNV

10-2

8.0 x 105 8/8

5.3 ±

0.3

10-4.5

4.4 x 103

10-3

8.0 x 104 8/8

5.3 ±

0.3

10-4

8.0 x 103 8/8

8.3 ±

2.0

10-5

8.0 x 102 0/8 -

10-6

8.0 x 101 0/8 -

Mice were monitored for 30 days for signs of disease and those that were sick were

sacrificed before they reached moribund state and deaths were recorded. The fifty

percent lethality dose (LD50) values were calculated according to the methods by Reed

and Muench (1938).

aInfectious units

bAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

84

Table 4.1.B. Mortality in different susceptible mouse strains following

intracerebral infection with KUNV.

Virus

Virus

stock

dilutions

Amount

(i.u.)a

%Mortality

(No. of died/

Inoculated)

ATDb

HeJ

10-2

3.0 x 104 85

(6/7) 6.0 ± 1.6

10-3

3.0 x 103

0

0/7 -

C57BL/6

10-2

3.0 x 104 64

(7/11) 5.8 ± 0.7

10-3

3.0 x 103

30

(3/10) 7.0 ± 2.0

BALB/c

10-2

3.0 x 104 90

(9/10) 5.6 ± 1.0

10-3

3.0 x 103

11

(1/9)

7.0 ± 0.0

aInfectious units

bAverage time to death (days) ± standard error

Differences in mortality between different mouse strains were not significant (p < 0.05)

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

85

4.2.1.2 Analysis of neurovirulence of WNV, KUNV and MVEV in resistant mice

Previous studies have indicated that the Flv gene does not confer full protection to

flavivirus resistant mice against all flavivirus infections (Sabin, 1954; Urosevic and

Shellam, 2002; reviewed in Brinton and Perelygin, 2004). Because of this, this study

was aimed at determining a difference in neurovirulence between three flaviviruses,

WNV, KUNV and MVEV in resistant DUB mice. As shown in Table 4.1.C,

intracerebral challenge of WNV and KUNV induced fatal encephalitis in a proportion of

resistant DUB mice. Parallel to infection in HeJ mice, KUNV induced a higher

death rate at a much lower dose than WNV. At 3.0 x 105 i.u., all DUB mice

succumbed to KUNV infection while none of the mice were affected following

challenge with equal to or less than 3.0 x 103 i.u. of KUNV. In contrast, WNV infection

at doses of 8.0 x 104 i.u. and 8.0 x 10

5 i.u., resulted in only 75% deaths of DUB mice.

However, the ATD in resistant DUB mice receiving various doses of WNV and KUNV

were considerably greater than that observed in similarly infected susceptible HeJ mice

(Student t test, p < 0.001), most probably due to the presence of the resistance gene in

the former mice that delayed a development of disease. More importantly, it was

demonstrated here that MVEV, which showed greater neurovirulence to HeJ mice than

KUNV and WNV, did not induce a fatal disease in DUB mice when inoculated at a very

high dose. However, higher dilutions of MVEV stock containing lower viral doses were

not included in this study since numerous studies previously performed in this

laboratory using lower doses of MVEV did not show any mortality among resistant

DUB mice (pers. com. Dr. N. Urosevic).

4.2.1.3 Analysis of different degrees of neurovirulence of WNV, KUNV and MVEV

In order to assess the neurovirulence levels of these three viruses, the virus dose

required to kill 50% of infected mice (LD50) was calculated (Reed and Muench, 1938).

From Table 4.1.A, LD50 in susceptible mice were 4.4 x 103 i.u., 1.7 x 10

3 i.u. and 3.4 x

101 i.u. for WNV, KUNV and MVEV respectively. From this result it can be concluded

that MVEV was the most neurovirulent virus in susceptible HeJ mice since fewer

infectious viral units were required to kill the same number of mice. The next most

neurovirulent virus was KUNV while WNV was the least neurovirulent virus in this

study (Table 4.1.A).

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

86

Table 4.1.C Mortality and LD50 studies following intracerebral infection with

serially diluted viruses in flavivirus resistant DUB mice.

Virus Virus stock

dilutions

Amount

(i.u.)a

Mortality/

tested ATD

b

LD50

(virus

dilutions)

LD50

(i.u.)

KUNV

10-1

3.0 x 105 10/10 8 ± 0

10-2.2

2.2 x

104

10-2

3.0 x 104

6/9 9 ± 0

10-3

3.0 x 103 0/10 -

10-4

3.0 x 102 0/10 -

10-5

3.0 x 101 0/10 -

MVEV* 10-1

1.0 x 105 0/10 - -

WNV

10-2

8.0 x 105 6/8 8 ± 0

10-3.5

6.2 x

104

10-3

8.0 x 104 6/8 9 ± 0

10-4

8.0 x 103 2/8 14 ± 0

10-5

8.0 x 102 2/8 14 ± 0

Mice were monitored for 30 days for development of diseases and those that were sick

were sacrificed before they reached moribund state and deaths were recorded. The fifty

percent lethality dose (LD50) values were calculated according to the methods by Reed

and Muench (1938). *Only one dose of MVEV was used to infect DUB mice since

numerous studies using equal or lower doses have been performed in this laboratory by

other researchers with similar outcome of infection observed (Silvia, 1999; Pantelic,

2004).

aInfectious units

bAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

87

Neurovirulence of these three flaviviruses in resistant DUB mice however was

significantly different. As demonstrated in Table 4.1.C, KUNV was the most

neurovirulence (LD50 was 2.2 x 104i.u.)

in resistant mice. WNV was slightly less

neurovirulent than KUNV since it had a higher LD50 value (LD50 6.2 x 104i.u.).

4.2.1.3 Mouse mortality and average time to death using a 100 LD50 virus dose

In this study, neurovirulence of the same flaviviruses was studied using single virus

dose of 100 times LD50 as determined for susceptible mice. This dose was known to

induce 100% mortality in susceptible mice, but the death rate in resistant mice was

unknown. Since in the previous studies (Section 4.2.1.3), KUNV and MVEV were

determined to be the most neurovirulent in susceptible and resistant mice, respectively,

they were chosen for further analyses of mortality rate and ATD in mice. A dose of 100

LD50 for susceptible mice corresponded to 1.74 x 105 i.u. of KUNV and 3.4 x 10

3 i.u. of

MVEV.

Two groups of HeJ mice and another two groups of DUB mice which consisted of 15

nine-week old animals per group were used for this experiment. One group of mice of

each mouse strain was challenged i.c. with KUNV while the other group with MVEV.

The mice were monitored for 30 days for the development of disease. Flavivirus

susceptible HeJ mice infected with KUNV started to show signs of sickness such as

slight fur ruffling and hunching four days after infection. The following day (day 5 p.i.),

all mice developed severe ruffling and hunching with some suffering from hind-leg

paralysis and they were immediately culled (Figure 4.1). In contrast, MVEV-infected

susceptible HeJ mice only started to exhibit signs of disease on day 5 p.i. and by day 6

p.i., all of them developed fatal encephalitis and were culled (100% mortality).

Similarly, KUNV infection in resistant DUB mice displayed a slower disease

progression compared with susceptible mice. Resistant DUB mice started to be slightly

ruffled and hunched on day 7 p.i. and subsequently all succumbed to the infection. Sixty

percent of DUB mice died on day 9 p.i. while the rest died the next day. In contrast,

resistant DUB mice challenged with MVEV did not develop any disease or death, as

shown previously in the laboratory (pers. com. Dr. N. Urosevic).

Therefore, using a dose of 100 LD50 for susceptible mice, earlier deaths were observed

in KUNV-infected susceptible HeJ mice (day 5 p.i.) than in those infected with MVEV

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

88

(day p.i.). In contrast, resistant DUB mice succumbed to KUNV infection 4-5 days later

(day 9 p.i.) while as expected, MVEV infection did not have any effect on resistant

mice.

4.2.2 INTRANASAL INFECTION IN SUSCEPTIBLE MICE

Intranasal infection (i.n.) of KUNV and MVEV was also performed in susceptible HeJ

mice to study mortality induced following an alternative route that delivers the virus

directly into the brain. This route avoids any needle injury to the brain that is observed

following i.c. inoculation. The injury caused to the brain following virus delivery during

i.c. infection may possibly contribute to the development of a more severe course of

infection. Thus, the mortality rate following i.n. challenge may be lower than that

observed after i.c. infection. In this study, four times the dose given for i.c. infection

was used for virus inoculation via i.n. route. A greater amount of virus was needed for

i.n. infection because it was known that a large percentage of the virus would be

distributed to the lungs, stomach or expelled from the nares of the animal (Silvia, 1999).

To study whether brain injury induced would exaggerate the course of infection, 5 µL

PBS was delivered i.c. into another group of mice a day after i.n. KUNV or MVEV

infection.

As shown in Table 4.2, mice that received i.c. inoculation of PBS only did not

succumbed to needle injury, suggesting that inflicted wound in the brain alone is not

sufficient to cause mortality in mice (Table 4.2). Only one of six susceptible HeJ mice

died from i.n. KUNV challenge while none of the 10 mice i.n. infected with MVEV

succumbed to the infection. Interestingly, the presence of brain wound increased

susceptibility of susceptible HeJ mice to i.n. KUNV and MVEV infection. Three out of

seven mice challenged with KUNV i.n. and had brain injury succumbed to the infection.

The difference however was not significant when compared to KUNV-infected mice

that did not receive PBS. In addition, the time to death and average brain viral burden

were similar to that observed in mice that received KUNV only. Following MVEV

challenge and brain injury, four out of ten mice showed serious signs of illness on day

10 p.i. and later died on day 11 or 12 p.i. (Student t test, p < 0.04 when compared with

infected with MVEV only).

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

89

0

20

40

60

80

100

1 3 5 7 9 11 13 15

Days post infection

Su

rviv

al

(%)

HeJ-KUNV HeJ-MVEV

DUB-KUNV DUB-MVEV

Figure 4.1 Analysis of survival in mice following infection with 100 LD50 (in

susceptible mice) of KUNV and MVEV.

The 100 LD50 dose of virus given to mice was equivalent to 1.74 x 105 and 3.4 x 10

3i.u.

of KUNV and MVEV, respectively. Fifteen mice were used per group and animals were

monitored until 30 days p.i. for signs of diseases.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

90

Table 4.2. Mortality studies following intranasal infection of KUNV and MVEV in

HeJ mice.

Virus

(i.n.)

Treatment

(i.c.)

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10 TCID50

/0.01g)

- PBS 0

(0/5) - -

KUNV

- 17

(1/6) 7.0 ± 0.0 7.4 ± 0.0

PBS 43

(3/7) 7.0 ± 0.0 7.7 ± 0.2

MVEV

- 0

(0/10) - -

PBS 40

(4/10) 11.8 ± 0.3 ND

The amount of virus given to the mice was 4 x 100 LD50. The values were equivalent to

6.9 x 105 i.u. and 1.4 x 10

4 i.u. of KUNV and MVEV respectively. Five microlitres of

PBS was i.c. injected a day after virus infection.

aAverage time to death (days) ± standard error

ND = not done

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

91

4.2.3 VIRUS NEUROINVASIVENESS STUDIES

4.2.3.1 Intraperitoneal challenge in adult and young mice

Neuroinvasiveness is the ability of certain viruses to replicate in the periphery and

invade the central nervous system to establish a disease. This property usually can be

tested by inoculating the virus from the periphery. Viruses that could cause morbidity in

mice following i.p. route of infection are classified as neuroinvasive viruses. Since

KUNV, MVEV and WNV have different neurovirulence properties in susceptible and

resistant mice, these flaviviruses were expected to display varying patterns or levels of

neuroinvasiveness as well.

To study neuroinvasiveness of KUNV, MVEV and WNV, three groups of adult 8 to 10

weeks old susceptible HeJ mice were infected i.p. with 2 x 107 i.u. of WNV, KUNV or

MVEV. This dose was chosen because it has been routinely used in the laboratory to

induce deaths in HeJ mice after i.p. challenge with WNV. As shown in Table 4.3,

infection with WNV resulted in 60% death of adult HeJ mice with ATD of about 9

days. The average brain titres in mice succumbing to WNV infection tested by TCID50

bioassay was 7.5 ± 0.5 log10 TCID50/0.01g brain tissue. Meanwhile, MVEV i.p.

challenge induced a slightly higher mortality rate (70%) in susceptible HeJ mice.

Furthermore, on average, susceptible HeJ mice succumbed to i.p. MVEV infection on

day 7.8 p.i., which was a day earlier than observed in WNV-infected mice (Student t

test, p > 0.05). Interestingly, brain viral titres of sick MVEV-infected HeJ mice were

more than 2 logs higher than in WNV-infected HeJ mice. In sharp contrast, KUNV

failed to induce fatal encephalitis in these mice. To further confirm that KUNV was

avirulent if inoculated by the peripheral route, animals were also challenged with higher

amounts of the virus. However i.p. challenge with 2 x 108 i.u. and 1 x 10

9 i.u. of KUNV

also did not induce any manifestation of disease in susceptible HeJ mice (data not

shown). Thus, it can be concluded that within the dosage range used in this study, only

WNV and MVEV were neuroinvasive while KUNV was not neuroinvasive in adult

flavivirus susceptible HeJ mice.

Since KUNV was non neuroinvasive in adult HeJ mice, a study was then performed to

look at the course of infection with the same virus in young 3-week-old mice. Young

mice have immature immune systems and not fully developed BBB and many studies

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

92

have shown that they are more susceptible to virus infection than adult mice (Liebert,

2001). Five young susceptible HeJ mice were given 2 x 107 i.u. of KUNV and

development of disease was monitored. Four animals started to develop ruffled fur and

hunched back 6 days after infection and by day 7 p.i., two of them exhibited hind-leg

paralysis and were sacrificed. Additionally, 2 mice succumbed to the virus infection on

day 9 p.i. Average brain virus titres from these dying mice were 5.84 ± 1.0 log10

TCID50/0.01g tissue (Table 4.3). This suggests that, in contrast to adult HeJ mice,

KUNV was highly neuroinvasive in young HeJ mice.

A similar neuroinvasiveness study was also undertaken in flavivirus resistant DUB

mice. However, in this part of the study, only WNV and KUNV were tested. MVEV

was excluded as it failed to induce any mortality in DUB mice even when virus was

directly inoculated into the brain (i.c. infection) (Section 4.2.1.2). It should be noted

here that the highest dose given to resistant DUB mice during i.c. challenge of MVEV

was 1 x 105 i.u., which was more than 0.5 logs higher than the amount of infectious

WNV and KUNV required to cause 50% mortality of DUB mice. It is not known

whether resistant DUB mice would still survive if a much higher dose of MVEV (more

than 1 x 105 i.u.) was used. However, it was impractical to infect resistant DUB mice

with more than 1 x 105 i.u. of MVEV as this would lead to inoculating virus at a neat

concentration since MVEV stock had a low TCID50 units compared to the other two

viruses. Given that virus was propagated in Vero cells, concentrated virus may contain

unnecessary components of the cells that could cause false positive/negative results

following infection.

When adult DUB mice received 2 x 107 i.u. of WNV or KUNV by the i.p. route (10

mice per virus), they did not show any signs of disease when monitored for 30 days

(Table 4.3). Similar infection in young 3 weeks old DUB mice (10 mice per virus)

produced comparable results (Table 4.3).

Since WNV was more neuroinvasive than KUNV in susceptible HeJ mice, higher

amounts of the former virus were also given to a group of young resistant DUB mice to

further verify that this virus was non neuroinvasive in resistant mice. Doses of 2 x 108

and 2 x 109 i.u. of WNV were given to 5 mice per group and this infection also failed to

induce lethal disease (data not shown). Thus, this study demonstrated that WNV and

KUNV were avirulent during i.p. challenge in both adult and young resistant DUB mice

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

93

Table 4.3. Intraperitoneal infection of KUNV, MVEV and WNV in flavivirus

susceptible HeJ and resistant DUB mice

Mouse

strain Virus

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10 TCID50

/0.01g)

HeJ

(adult)

WNV 60

(6/10) 9.0 ± 0.8 7.5 ± 0.5

KUNV 0

(0/15) - -

MVEV* 70

(7/10) 7.8 ± 0.5 10.0 ± 0.3

HeJ

(young) KUNV

80

(4/5) 8.0 ± 1.0 5.84 ± 1.0

DUB

(adult)

WNV 0

(0/10) - -

KUNV 0

(0/10) - -

DUB

(young)

WNV 0

(0/10) - -

KUNV 0

(0/10) - -

Adult mice used were aged between 8 to 12 weeks old while young mice were aged

between 3 to 4 weeks old, and they were infected i.p. with 2 x 107 i.u. of virus.

*Since MVEV was not virulent to resistant mice following i.c. inoculation, it was

excluded from the neuroinvasiveness studies.

aAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

94

which most probably due to the flavivirus resistance gene that confers resistance to

peripheral tissues, consequently preventing virus replication and subsequent viral

invasion of the CNS.

4.2.3.2 Effect of blood brain barrier modulation on virus neuroinvasiveness

Following natural infection, flaviviruses are thought to spread, at least in part, via the

haematogenous route (Chambers and Diamond, 2003). For many encephalitis-inducing

viruses, the major obstacle is to pass the BBB, which forms a tight junction of the

endothelial cells in the brain and controls influx of molecules into the CNS. Studies

have shown that if the permeability of BBB is breached, non-neuroinvasive viruses may

also be able to reach the brain (Lustig et al, 1992; Kobiler et al, 1989; Ben-Nathan et al,

1996). Thus, it would be interesting to investigate whether increasing permeability of

the BBB would alter the outcome of infection during i.p. challenge of KUNV, MVEV

and WNV in susceptible and resistant mice.

4.2.3.2.1 Effect of SDS on KUNV and MVEV neuroinvasiveness in HeJ mice

The present study was designed to test whether SDS, which is known to modulate BBB

permeability (Saija et al, 1997), would increase susceptibility of HeJ mice to i.p. KUNV

and MVEV infections. Kobiler and co-workers (1989) showed that a variant of WNV-

25, that has lost neuroinvasiveness but not neurovirulence, could cause mortality in

mice when SDS (about 60 ng) was given together with the virus by the i.v. challenge.

Neuroinvasiveness of KUNV and MVEV were monitored in this study as these viruses

were shown to have very contrasting virulence traits in mice. While MVEV is highly

virulent in susceptible HeJ mice only, KUNV does not kill adult susceptible HeJ mice

by the i.p. route but it induces high morbidity in HeJ and DUB mice following i.c.

challenge.

To look at the ability of SDS to alter mortality in susceptible adult HeJ mice, a group of

15 mice were given SDS and KUNV at the same time (Table 4.4). Since several pilot

experiments performed by the i.v. route did not provide satisfactory results and due to a

limited number of animals available for optimising this experiment, the i.v. route of

inoculation was abandoned and the i.p. injection was chosen for SDS inoculation into

the mice. Two hundred microlitres of PBS that contained 240 ng SDS and 2 x 107 i.u.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

95

virus were given i.p. to a group of 10 to 15 adult HeJ mice and they were under

observation for 30 days. In addition, mice infected only with virus (control) were also

included. In KUNV-infected susceptible HeJ mice, concurrent administration of SDS

and virus did not have any effect on HeJ mice as all mice survived i.p. KUNV infection.

From the study above, SDS did not alter the survival rate of adult susceptible HeJ mice

when administered to the mice at the same time as the virus. It is possible that such an

outcome was due to a low efficiency of the i.p. challenge compared to the i.v.

inoculation, which used by Kobiler and co-workers (1989). Intravenous inoculation

transports virus directly into the bloodstream. In contrast, i.p. injection distributes virus

into the peritoneal cavity and possibly, only a small amount of virus could get into the

bloodstream, causing a delayed and low viraemia. Thus, the next set of experiments was

designed to treat mice with SDS several days after i.p. virus infection, allowing virus

replication and the induction of viraemia. Viremia was shown to occur up to day 3 p.i.

following i.p. infection with WNV in susceptible HeJ mice (Pantelic, 2004). However,

there was no prior knowledge on the levels and extent of viraemia during i.p. KUNV

and MVEV infection in this laboratory and thus it was decided that SDS would be given

3 days after KUNV or MVEV inoculation to adult HeJ mice. As illustrated in Table 4.4,

this treatment increased the susceptibility of adult HeJ mice to i.p. MVEV but not i.p.

KUNV challenge. During i.p. MVEV challenge in control mice, animals started to

exhibit typical signs of flavivirus-induced encephalitis including ruffled fur and

hunched posture on day 5 p.i. Deaths occurred between day 6 p.i. to day 9 p.i. and 70%

mortality was recorded (7 out 10 mice died). Average time to death of these mice was

7.8 days p.i. with average brain titres of 10.0 ± 0.3 log10 TCID50/0.01g tissue. In

contrast, SDS treatment 3 days after infection caused quicker and slightly higher rate of

lethal MVEV (80%, p > 0.05, Kaplan Meier test ) infection in susceptible HeJ mice.

The first death of MVEV-infected susceptible HeJ mice treated with SDS was recorded

on day 4 p.i., which was 2 days earlier than control mice (Table 4.4). On average, these

mice succumbed to fatal disease a day earlier than the control mice (6.8 days p.i.).

While brain viral burden was slightly lower than in non-SDS treated group, no

considerable difference was noted between these 2 groups of mice (Student t test, p >

0.05).

Since SDS treatment did not enhance susceptibility of adult HeJ mice to i.p. KUNV

challenge, a small scale study involving young HeJ mice was carried out. Five mice

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

96

were used per group and they either received virus only, virus and SDS concurrently or

virus and SDS 3 days p.i. As shown in Table 4.4, concurrent SDS and i.p. KUNV

inoculation did not increase vulnerability of young HeJ mice to KUNV infection

since similar rate of mortality (80%) and ATD ( 8-9 day p.i.) were recorded.

Meanwhile, SDS treatment 3 days after virus challenge exacerbated the course of i.p.

KUNV infection in young HeJ mice. Rate of fatal encephalitis increased from 80% to

100% although statistical analysis could not be performed due to the small number of

mice involved. The increased susceptibility to i.p. KUNV infection was further

evidenced by the rapid disease progression. Young HeJ mice started to develop disease

on day 4 p.i. and some deaths occurred on day 5 p.i., a day earlier than that observed in

mice not treated with SDS. On average, SDS-treated mice succumbed to the infection

on day 6 p.i while non-treated animals died on day 8 p.i. However, the average brain

viral titres in SDS-treated KUNV-infected mice were a log lower than observed in non-

SDS treated mice.

4.2.3.2.2 Effect of blood brain barrier modulation on WNV neuroinvasiveness in mice

It remained to be answered whether resistant DUB mice would exhibit similar increased

susceptibility to i.p. flavivirus infection following breaching of the BBB. However,

since KUNV is a non neuroinvasive virus in adult susceptible mice while MVEV is

avirulent in resistant mice, WNV was the best alternative virus for neuroinvasiveness

study in resistant DUB mice since this virus is highly virulent to HeJ and DUB mice.

The effect of SDS on the neuroinvasiveness of WNV was investigated in young and

adult resistant DUB mice. In this study, adult HeJ mice were also included as a control

and since contribution of SDS to the outcome of i.p. WNV infection in susceptible mice

was not known. Mice were either infected with virus only or virus and SDS on either

day 0 or 2 p.i. Day 2 p.i. was chosen in this particular because it was thought that if

DUB mice developed viremia following i.p. flavivirus infection, it would be a much

transient process than that observed in HeJ mice. Additionally, day 2 p.i. corresponded

to the peak of viremia following the i.p. challenge with WNV.

As expected, concurrent administration of SDS and virus did not modify the survival

rate of adult HeJ mice as 60% of mice died with comparable ATD and average brain

viral titres (Student t test, p > 0.05) when the virus was i.p. inoculated with or without

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

97

SDS (Table 4.5). Meanwhile, WNV-infected HeJ mice treated with SDS 2 days after

infection exhibited higher susceptibility to the virus. Mortality increased to 75%

following SDS treatment although this was not markedly different. SDS treatment

however did not alter the resistance of DUB mice to i.p. WNV challenge (Table 4.5).

In addition to SDS, resistant mice were also treated with LPS during or after i.p. WNV

infection. LPS is an outer component of the cell wall of some gram negative bacteria

and can cause an array of physiological changes including immuno-stimulatory and

antigenic responses, and is known to alter BBB permeability (Di Marzio et al, 1990).

Lustig and co-workers (1992) previously demonstrated that administration of LPS prior

to infection with a non-neuroinvasive variant of WNV caused some mortality in mice.

While the majority of mouse strains respond to LPS, some mouse strains including HeJ

mice do not (Silvia and Urosevic, 1999). Resistant DUB mice however, were shown by

Silvia and Urosevic (1999) to be an LPS-responsive mouse strain since LPS-responsive

C3H/HeJARC mice were used as inbred parents during production of congenic resistant

DUB mice. Due to the LPS unresponsiveness of the HeJ mice, the effect of LPS on

modulation of WNV neuroinvasiveness was only studied in resistant DUB mice (data

not shown). LPS can stimulate macrophages and B cells to produce cytokines including

IL-1, TNFα and IFNγ (Di Marzio et al, 1990; Cockfield et al, 1993; Lustig et al, 1992).

Thus, to further verify the response of DUB mice to LPS, the levels of sera TNFα were

measured using a commercial ELISA-based assay (eBioscience). Sera were collected

from 2 non-treated mice and 2 LPS-treated mice (1.5h after treatment at 50 μg/mouse).

While TNFα was below detection in non-treated mice, LPS-treated mice had average

sera levels of 14.25 ± 7.5 pg/mL TNFα (data not shown), further confirming the

responsiveness of DUB mice to LPS.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

98

Table 4.4 The effect of SDS on mortality of HeJ mice following i.p. KUNV and

MVEV infection.

Virus Age of

mice Treatment

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10

TCID50

/0.01g)

- Adult SDS 0

(0/5) - -

KUNV Adult

- 0

(0/10) - -

SDS

(day 0 p.i.)

0

(0/15) - -

SDS

(day 3 p.i.)

0

(0/10) - -

KUNV Young

- 80

(4/5) 8.0 ± 1.0 5.8 ± 1.0

SDS

(day 0 p.i.)

0

(4/5) 9.0 ± 1.0 5.9 ± 1.0

SDS

(day 3 p.i.)

100

(5/5) 6.0 ± 1.0 4.9 ± 1.1

MVEV Adult

- 70

(7/10) 7.8 ± 0.5 10.0 ± 0.3

SDS

(day 3 p.i.)

80

(8/10) 6.8 ± 0.5 9.2 ± 0.7

Mice received 2 x 107 i.u. of virus and 240 ng of SDS. The animals were monitored for

30 days for any signs of diseases.

aAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

99

However, when LPS was administered to young and adult resistant DUB mice at the

same dose as above (50 μg/mouse i.p.) immediately or 2 days after i.p. WNV infection,

no development of fatal encephalitis was observed in these mice (data not shown). The

lack of effect of LPS on WNV neuroinvasiveness is not clear since no study was

performed to monitor the breaching of the BBB following treatment with 50 μg LPS.

It is possible that the lack of severe WNV infection in LPS-treated mice was due to

inefficient LPS treatment on the BBB permeability or the lack of peripheral virus

replication which eventually did not allow viraemia and viral invasion of the CNS.

Collectively, data from this study suggest that increasing the BBB permeability 2 to 3

days after infection only enhanced susceptibility of HeJ mice but not DUB mice to i.p.

flavivirus challenge, evidenced by the shorter ATD and/or increased death rate in the

former mice, although this depends on the strains of infecting virus as well.

4.2.3.3 Effect of macrophage modulation on flavivirus infection

Macrophages have pivotal role in the host antiviral immunity. Macrophages can act as

APC and produce various cytokines that could contribute to the clearance of virus

infection (Beutler, 2004). However, the ability of monocytes and macrophages to

support flaviviruses has been reported and therefore they may be also involved in the

pathogenesis of some viruses including DENV (reviewed in Solomon and Mallewa,

2001). In this part of study, the role of macrophages in immunopathogenesis and host

defence during flavivirus infection in both susceptible and resistant mice was

investigated as well as whether the modulation of macrophages using selected

chemicals would affect mortality or survival of mice after flavivirus infection was

investigated.

4.2.3.3.1 Mouse survival following thioglycollate treatment

The objective of this study was to investigate whether manipulation of macrophages

using thioglycollate could modify the outcomes of WNV and KUN infections in

flavivirus susceptible and resistant mice. Thioglycollate is a sterile inflammatory agent

that has been used extensively in many laboratories to attract cells into the peritoneal

cavity. This treatment has been shown previously to cause up to 10-fold increase in the

number of peritoneal cell yields with macrophages comprising more than 80% of these

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

100

cells (Silvia et al, 2001). To investigate the effect of thioglycollate treatment on

flavivirus infection, 10 adult HeJ mice were infected i.p. with 2 x 107 i.u. of WNV or

KUNV. In another two groups of mice (10 mice per group), 1mL of 6% thioglycollate

was given i.p. to mice 3 days prior to WNV or KUNV infections. As shown in Table

4.6, accumulation of macrophages in the peritoneal cavity increased susceptibility of

adult HeJ mice to i.p. challenge with WNV, indicated by the increased death rate from

60% to 100% (Kaplan Meier test, p < 0.03). This result was in agreement with the

previous findings in this laboratory (Silvia, 1999; Pantelic, 2004). These mice started to

become sick on day 6 p.i. and the disease progressed in the next few days.

Most of the deaths were recorded on day 8 p.i. In contrast, WNV-infected mice not

treated with thioglycollate did not succumb to fatal encephalitis until 9 days after

infection. This indicates that peritoneal macrophages may be involved in WNV

pathogenesis and consequently in a fatal disease outcome, possibly by increasing virus

dissemination. Surprisingly, thioglycollate did not affect the survival of KUNV-infected

HeJ mice. The resistance of WNV infected adult and young DUB mice was similarly

unaffected by this chemical (Table 4.6).

Another separate group of young and adult resistant DUB mice were treated with

thioglycollate 3 days prior to infection and later with SDS 2 days after i.p. WNV

infection. It is initially thought that these treatments would make DUB mice more

vulnerable as these chemicals not only elicited potential harbouring site in the peritoneal

cavity but at the same time may assist WNV to invade the brain by breaching the BBB.

However, as shown in Table 4.7, DUB mice did not show increased susceptibility to i.p.

WNV challenge following both thioglycollate and SDS treatments.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

101

Table 4.5 The effect of SDS on mortality of mice following i.p. WNV infection in

mice.

Mice Treatment

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10

TCID50

/0.01g)

HeJ

(adult)

- 60

(6/10) 9.0 ± 1.0 7.5 ± 0.5

SDS

(day 0 p.i.)

60

(6/10) 10.0 ± 1.0 6.9 ± 0.3

SDS

(day 2 p.i.)

75

(6/8) 9.0 ± 0.5 ND

DUB

(adult)

- 0

(0/10) - -

SDS

(day 0 p.i.)

0

(0/13) - -

SDS

(day 2 p.i.)

0

(0/8) - -

DUB

(young)

- 0

(0/10) - -

SDS

(day 0 p.i.)

0

(0/10) - -

SDS

(day 2 p.i.)

0

(0/5) - -

Mice were infected with 2 x 107 i.u. of virus and received either 240 ng of SDS or 50 μg

LPS. The animals were monitored for 30 days for any signs of disease. Administration

of only SDS in DUB mice did not result in development of disease.

aAverage time to death (days) ± standard error

ND = not done

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

102

4.2.3.3.2 Mouse survival following transient macrophage depletion

Another important role played by macrophages is to provide an early non-specific

defence against invading micro-organisms. Thus, to study the importance of

macrophages in the early protection against flavivirus infection in susceptible and

resistant mice, transient depletion of macrophages was performed prior to virus

infection. Selective depletion of macrophages can be achieved by local administration

of liposome encapsulated dichloromethylene bisphosphonate (clodronate).

Intraperitoneal inoculation of clodronate depletes macrophages in the peritoneal cavity,

spleen, parathymic lymph nodes and liver (van Rooijen and Sanders, 1994; Biewenga et

al, 1995; Ciavarra et al, 1997). Five adult mice of each HeJ and DUB strains were

treated i.p. with 100μL of clodronate liposomes. Another 10 HeJ mice were treated with

100μL of empty liposomes. Four days later, all of these mice were infected i.p. with

WNV at 2 x 107 i.u./mouse and they were monitored for 30 days. Control mice infected

with WNV only were also included. The depleting effect of clodronate on tissue

macrophages was confirmed by taking spleens from control and clodronate-treated HeJ

mice (4 days after treatment) and staining macrophages for F4/80 cell surface antigen

on the frozen spleen sections (Figure 4.2). Clodronate administration was shown to

cause elimination of the marginal zone and the red pulp macrophages from spleens of

treated mice. However, it must be noted here that macrophage depletion by clodronate

is a transient process. Since clodronate was administered only once, the absence of

macrophages was certain only at the early stage of infection and not throughout the

course of infection.

As shown in Table 4.7, HeJ mice that received empty liposome had similar mortality

rate and ATD as the control WNV-infected mice. This shows that phagocytosis of

empty liposomes did not affect WNV pathogenesis. Interestingly, mortality increased

from 60% in the control group to 100% in the clodronate-treated group (Table 4.7).

However, the overall statistical significance could not be calculated due to the small

numbers of animal used. Disease progression was notably quicker in clodronate-treated

mice which became sick on day 4 pi and all died the following day (Student t test, p <

0.05). In contrast, control HeJ mice only started to succumb to i.p. WNV from day 8 p.i.

onwards. Clodronate-treated HeJ mice succumbing to the i.p. WNV challenge also

displayed about 2.5 log lower brain virus titres than control HeJ mice (Student t test, p <

0.05). Thus, the transient absence of macrophages had a significant effect on i.p. WNV

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

103

Table 4.6. The effect of thioglycollate on mortality of HeJ and DUB mice following

i.p. virus infection.

Mouse

strain Virus Treatment

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days

p.i.)

Viral titres

(log10 TCID50

/0.01g)

HeJ

(adult)

WNV

- 60

(6/10)

10.0 ±

2.0 7.5 ± 0.5

Thioglycollate 100

(10/10) 8.0 ± 1.0 7.0 ± 1.0

KUNV

- 0

(0/10) - -

Thioglycollate 0

(0/10) - -

DUB

(adult) WNV

- 0

(0/5) - -

Thioglycollate 0

(0/5) - -

Thioglycollate

/SDS (day 2

p.i)

0

(0/9) - -

DUB

(young) WNV

Thioglycollate 0

(0/13) - -

Thioglycollate

/ SDS (day 2

p.i.)

0

(0/6) - -

Adult mice were given 1mL of 6% thioglycollate and 3 days later received 2 x 107 i.u.

of virus. Two hundred microlitres of thioglycollate was administered to young DUB.

The animals were monitored for 30 days for any signs of disease.

aAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

104

Table 4.7. The effect of macrophage depletion on mortality of adult HeJ and DUB

mice following WNV infection

Mouse

strain Treatment

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10 TCID50

/0.01g)

HeJ

- 57

(4/7) 8.5 ± 1.1 6.8 ± 0.4

Empty

liposome

60

(6/10) 8.3 ± 1.0 7.0 ± 0.5

Clodronate 100

(5/5) 5.0 ± 0.0* 5.4 ± 1.6*

DUB Clodronate 0

(0/5) - -

Clodronate (100 μL) was given to the mice 4 days prior to WNV infection at

2 x 107 i.u. The animals were monitored for 30 days for any signs of disease. Statistical

analysis was not performed due to the small numbers of mice involved in this study.

*The difference in the ATD and brain viral titres were statistically significant in

clodronate treated-HeJ versus control or liposome treated-HeJ mice (Student t test, p <

0.05)

aAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

105

A

B

Figure 4.2: Depletion of splenic macrophages by clodronate treatment.

Frozen spleen tissue sections taken from mice 4 days after receiving 100μL of

A) PBS B) Clodronate. Macrophages were stained with monoclonal antibody specific

for F4/80 cell surface antigen which is expressed in macrophages (brown colour). No

splenic macrophages could be detected in mice treated with clodronate.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

106

infection in HeJ mice, suggesting that these cells may have important role in the early

host innate defence against WNV.

However, similar to earlier observations in BBB modulation studies, elimination of

macrophages early in the infection by clodronate did not promote development of fatal

disease in resistant DUB mice following i.p. WNV infection. These findings indicate

that the outcome of infection varies in different animal models and following different

treatments since virus pathogenesis is dependent on many host and viral factors.

4.2.3.4 Effect of T cells depletion on survival of DUB mice following WNV i.p.

infection

An intact host immune response is critical for prevention of fatal diseases following

invasion of pathogens (Chambers and Diamond, 2003; Diamond, 2003). In addition to

the host innate defence mechanisms/cells, the host adaptive immune response has been

shown to have a crucial role in the protection or pathogenesis of some flaviviruses,

depending on the infecting flaviviruses, route of inoculation as well as the genetic

background of the mice (reviewed in Chambers and Diamond, 2003; King et al, 2003;

Wang et al, 2003b). Thus, the following study was designed to examine the role of T

cells, which are part of the host adaptive immune cells, in resistant mice during i.p.

WNV challenge. The depletion effect of CD4+ T cells and/or CD8+ T cells was

achieved using anti-CD4+ T cells or CD8+ T cells monoclonal antibodies. This study

was performed in resistant DUB mice only due to limited amount of depleting

monoclonal antibodies available. To confirm that depletion of T cells was achieved, two

groups of mice were given either anti-CD4+ or anti-CD8+ T cell depleting antibodies

on day 1 and day 3. Spleens harvested from these mice on day 4 post depletion were

then analysed by flow cytometry. 96% and 95% depletion of CD4+ and CD8+ T cells

was achieved, respectively (please refer to Figure 6.5).

Young 3 week old DUB mice were depleted of CD4+ T cells (150 µL) while adult DUB

mice were depleted of both CD4+ (150 µL) and CD8+ T (100 µL) two days prior to i.p.

challenge with WNV. Since 2 x 107 i.u. of WNV did not kill DUB mice by i.p.

challenge, a higher dose of 2 x 108 i.u. was used in this study. It was thought that this

high viral dose would be sufficient to induce fatal encephalitis in resistant DUB mice

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

107

with a breached immune response. Following infection, animals received further T cell-

depleting antibodies on day 0, 2, 4 and 8 p.i.

However, as shown in Table 4.8, absence of CD4+ T cells in young 3 weeks old DUB

mice as well as the lack of both CD4+ and CD8+ T cells in adult DUB mice did not

render these animals susceptible to i.p. WNV infection when monitored for 30 days.

This suggests that the flavivirus resistant gene has a more dominant role in determining

the outcome of infection than other forms of host non-specific or specific immune

responses.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

108

Table 4.8. The effect of T cells depletion on mortality of DUB mice following i.p.

WNV infection

Age of

mice

(weeks)

T cells

depletion

Mortality (%)

(number of

death/total

number of mice)

ATDa

(days p.i.)

Viral titres

(log10 TCID50

/0.01g)

3 - 0

(0/5) - -

3 CD4 0

(0/5) - -

7 CD4/CD8 0

(0/5) - -

Resistant mice were infected with 2 x 108 i.u. of WNV and depletion was performed

on 2 days before infection, day 0, 2, 6, and 8 p.i. Mice were monitored for 30 days for

any signs of illness.

aAverage time to death (days) ± standard error

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

109

4.3 DISCUSSION

This study described the in vivo characterisation of three closely related flaviviruses,

KUNV, MVEV and WNV in flavivirus susceptible HeJ and resistant DUB mice. The

most important finding of this study was that different virulence traits of KUNV,

MVEV and WNV were observed in these two mouse strains differing in their resistance

to flaviviruses. These results are remarkable as they further support other previously

published reports on flavivirus pathogenesis which involves multifaceted processes and

in which the outcome of infection is virus type- and host-dependent. Even closely

related flaviviruses such as WNV and KUNV behave differently in the same mouse

strain. In susceptible HeJ mice, KUNV, MVEV and WNV were neurovirulent, although

different degrees of neurovirulence were exhibited by these three viruses. MVEV was

the most neurovirulent virus in these susceptible mice, followed by KUNV and WNV,

as demonstrated by the low amount of MVEV (LD50 was 3.4 x 10 i.u.) needed to kill

50% of susceptible mice, compared with LD50 of 1.7 x 103 i.u. for KUNV and LD50 of

4.4 x 103 i.u. for WNV (Table 4.1.A). Based on the LD50 in HeJ mice following i.c.

inoculation as shown above, KUNV appears to be significantly more virulent than

WNV (one log difference in the LD50). However, as it will be discussed later, WNV

was neuroinvasive in HeJ mice following i.p. inoculation while KUNV was not. These

interesting findings suggest that the virus virulence is a very complex property and it

depends on the route of virus inoculation and the genetic background of the host.

Until now, there was no evidence to suggest that there are different levels of

susceptibility to flaviviruses among laboratory mouse strains carrying Flvs gene

(Darnell et al, 1974; Sangster et al, 1993). Although Muira and colleagues (1988)

reported a variable susceptibility of C3H, C57BL/6 and BALB/c mice following i.p.

challenge with JEV, these differences were not observed when mice were infected with

JEV via the i.c. route. Accordingly, in this current study, HeJ, C57BL/6 and BALB/c

mice which have different genetic backgrounds were also shown to be equally

susceptible to i.c. KUNV challenge with comparable mortality rates and ATD observed

(Table 4.1.B). This is the first time that the i.c. infection with KUNV strain MRM16

was studied in parallel in mice of three different flavivirus susceptible mouse strains,

revealing a similar neurovirulence of KUNV in these mice.

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

110

Intracerebral infection of the same viruses in resistant DUB mice yielded very distinct

outcomes of infection compared to susceptible HeJ mice. Interestingly, KUNV was the

most neurovirulent virus in resistant DUB mice, followed closely by WNV.

Remarkably, MVEV, which was highly virulent in HeJ mice, was not neurovirulent in

resistant DUB mice (Table 4.1.C). The disparity in the neurovirulence of MVEV in

flavivirus susceptible and resistant mice indicates a dominant contribution of the host

resistance factor, Flvr-like, to the outcome of infection. Another related host resistance

factor, Flvr, was also shown in other studies to confer strong protection to resistant RV

mice against infection with a number of flaviviruses (Urosevic et al, 1999; Urosevic and

Shellam, 2002; reviewed in Brinton and Perelygin, 2003). The genes controlling these

host resistance factors were mapped to mouse chromosome 5 (Sangster et al, 1999;

Urosevic et al, 1997b). Recently, a gene candidate for Flvr was identified as an OAS1b

gene (Brinton and Perelygin, 2003). The mice carrying the Flvr and Flv

r-like genes were

shown to be fully resistant to MVEV OR2 strain (Shueb et al, 2005; Urosevic et al,

1999). However, these mice succumbed to infection with WNV and KUNV following

i.c. inoculation (Shueb et al, 2005) and these findings were in agreement results from

the present study.

One interesting observation was that although resistant DUB mice succumbed to i.c.

WNV and KUNV infection, the effect of the Flvr-like gene was also evident, as

indicated by the longer ATD (between 8 to 14 days) in infected DUB mice compared to

susceptible HeJ mice (about 5 days). Furthermore, the viral dose required to kill 50% of

resistant mice was 25 and 14 times greater than that required to kill similar proportion

of susceptible mice following i.c. infection with KUNV and WNV, respectively. The

vulnerability of flavivirus resistant DUB mice to certain i.c. flavivirus infections was

seen not only in this study. In fact, resistant mice have been reported to die from i.c.

infection with certain strains of WNV, JEV, SLEV and MVEV (Sabin, 1952a; Sabin,

1952b; Urosevic and Shellam, 2002; Brinton, 2001). However, the molecular or

cellular mechanisms responsible for this phenomenon have never been investigated

before.

A separate experiment was performed in this study examining the possibility that an

injury caused by a needle while performing i.c. delivery of a virus could exaggerate the

severity of the diseases. Hase and co-workers (1990b) have demonstrated that needle

injury increased mortality rate as well as decreased survival time in mice i.p. infected

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

111

with JEV. Accordingly, it was demonstrated in this study that HeJ mice exhibited

enhanced susceptibility to i.n. KUNV and MVEV when a wound was inflicted in the

brain following infection. This might suggest that the fatal encephalitis observed in

mice challenged i.c. can be influenced by the needle injury during administration of

virus.

Further work on in vivo characterisation of flaviviruses in this study demonstrated that

WNV and MVEV were neuroinvasive in susceptible HeJ mouse strain. This was

reflected by the ability of WNV and MVEV to induce 60% and 70% mortality,

respectively, in these mice. Interestingly, MVEV was slightly more neuroinvasive than

WNV since it induced earlier manifestation of fatal encephalitis in HeJ mice than WNV

(ATD of day 7.8 compared to ATD of day 9 during MVEV and WNV infection,

respectively). In contrast, KUNV was non neuroinvasive and it did not promote

morbidity in adult HeJ mice even when they were infected with much higher doses; up

to 109 i.u. of KUNV. KUNV was recently classified as the lineage I WNV and

therefore it is closely related to virulent WNV strains that are associated with fatal

human encephalitis in Europe, Russia, North America and the Middle East (Hall et al,

2002). At the proteome level, KUNV is more than 98% homologous in amino acid

sequence to WNV NY99 strain (Liu et al, 2003; Shi et al, 2002). However, to date,

KUNV is considered to be a flavivirus with minor medical implications as infections in

humans with KUNV are mild, with clinical symptoms including mild fever, headache,

rash and myalgia (Hall et al, 2002). This is in agreement with the findings in this study

which demonstrated that KUNV was non-neuroinvasive in adult HeJ mice. In fact,

fatalities in humans have not been recorded since the discovery of KUNV more than 40

years ago (Hall et al, 2002). Furthermore, data attained in the present study are in

accordance with results reported by several other investigators regarding the inability of

KUNV to kill mice during peripheral infection (Scherret et al, 2001; Beasley et al,

2002).

Intraperitoneal infection, unlike intracerebral infection, results in a broad dissemination

of the virus throughout the body with only a small proportion of it directly reaching the

brain. This is in contrast to the i.c. challenge, where most of the virus is delivered

directly to the CNS. Neuroinvasive viruses usually possess the ability to replicate to

high titres in the peripheral tissues/organs and to promote high levels of viraemia that

subsequently facilitates viral invasion of the brain (reviewed in Chambers and

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

112

Diamond, 2003). Although investigation into infection and viraemia in peripheral

organs were not conducted in the current study due to limited animals available, it could

be hypothesised here that the profound difference in neuroinvasiveness between

MVEV/WNV and KUNV in susceptible adult HeJ mice may be caused by the increased

infectivity and tropism towards extraneural tissues of MVEV/WNV, enabling them to

replicate and multiply at higher titres than KUNV. This would lead to sufficient levels

of viraemia to promote viral invasion of the brain. An association between different

tropism for extraneural tissues and the ability of viruses to generate viraemia has been

reported previously in high and low neuroinvasive flaviviruses (McMinn et al, 1996). In

this study, the highly neuroinvasive MVEV strain replicated in the lymph node and

induced early viraemia while the low neuroinvasive variant did not. Thus, the observed

lack of neuroinvasiveness of KUNV in susceptible HeJ mice may be explained by the

inefficient extraneural tissue replication of this virus.

Although KUNV is known to be non-neuroinvasive in adult susceptible mice (Scherret

et al, 2001, Beasley et al, 2002), it was the first time that KUNV was shown to be

extremely neuroinvasive in young HeJ mice, as evidenced by the high fatal encephalitis

rate in these mice (Table 4.3). Again, this finding highlights the complex host-viral

interaction and that different dominant factors govern the severity of disease in different

infection model. In young HeJ mice, the increased susceptibility to i.p. KUNV was due

to various host factors, particularly the age-related susceptibility. These include the less

developed adaptive immune response, poorly developed apoptotic regulator, IFN-

responsive genes and others that may allow a greater level of peripheral viral replication

and elevated viraemia (reviewed in Mullbacher et al, 2003; Labrada et al, 2002).

Additionally, young mice possess immature BBB which facilitates virus spread into the

brain. In humans, young children have also been reported to be more prone to develop

encephalitis with poor prognosis, as well as having a greater chance of suffering from

neurological sequelae after virus infection in comparison to older children and adults

(reviewed in Griffin, 1995).

In this study, it was reported for the first time that KUNV and WNV, although highly

neurovirulent to resistant DUB mice, are not neuroinvasive in these mice as shown by

their inability to induce fatal encephalitis following i.p. inoculation despite their

neurovirulence in the same resistant mouse strain during i.c. infection (Table 4.3). In

case of WNV, the lack of neuroinvasion in resistant DUB mice may have resulted from

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

113

the expression of Flv-resistance in peripheral tissues. Expression of resistance has been

shown in primary cultures prepared from lungs, kidneys, spleen, peritoneal

macrophages and embryo fibroblasts from resistant mice (reviewed in Brinton and

Perelygin, 2003). Although flavivirus resistant cells can be infected, virus usually

replicates at low titres (Urosevic and Shellam, 2002). As a consequence, peripheral

organs probably were infected at a very low level with WNV, leading to insufficient

viraemia. Since KUNV was non-neuroinvasive to susceptible HeJ mice, its inability to

infect resistant DUB mice when inoculated peripherally is not surprising and clearly, it

is not dependent on the expression of the Flv resistance gene. However, the Flv gene is

known to be expressed even in young resistant mice (Sabin, 1952). In this study, it was

demonstrated that 3 weeks old DUB mice were also not susceptible to i.p. challenge

with either KUNV or WNV. Furthermore, these mice survived i.p. WNV infection at a

high dose (2 x 109 i.u.), suggesting that the effect of the Flv

r gene was dominant and

protective to young mice even in the absence of mature adaptive immunity.

The second aim of this study was to investigate the effect of modulation of the host

innate defence and adaptive immunity on flavivirus neuroinvasiveness in susceptible

HeJ and resistant DUB mice. The ability of flaviviruses to induce mortality in mice

following i.p. challenge depends on many host and viral factors including virus tropism

for peripheral tissues and its ability to cross the brain, as discussed previously. Some

physical alterations of the host such as breaching the BBB and immunosuppression

have been shown to result in the increased vulnerability of the host to viral challenge as

virus may have easier access to establish fatal infection (reviewed in Chambers and

Diamond, 2003; Kobiler et al, 1989; Lustig et al 1992; Saija et al, 1997). Thus, in this

part of the study, the correlation between increased BBB permeability by SDS and LPS,

absence and increased presence of peritoneal macrophages by clodronate and

thioglycollate, respectively, and transient loss of T cells with the severity of infection

and outcome of infection was examined.

SDS is a chemical that causes temporary and reversible breach of BBB (Saija et al,

1997). It has an amphiphilic property which enables this chemical to enter into

interactions with the major membrane components; lipids and proteins (Saija et al,

1997). Unfortunately, in the present work, there was insufficient time to optimise the

effect of SDS on the BBB in terms of the route of administration, (i.v. versus i.p.) and

the duration of the effect on the BBB. However, results obtained in this study following

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

114

administration of SDS two or three days after infection with WNV and KUNV showed

an increased incidence of fatal encephalitis in susceptible HeJ mice, as discussed below,

indicating that SDS has an effect on virus neuroinvasiveness.

Concurrent i.p. administration of virus and 240ng SDS did not enhance susceptibility of

adult HeJ mice to flaviviruses tested in this study (Table 4.4 and 4.5). However,

increased mortality rate and reduced ATD were observed when SDS was given two to

three days after i.p. MVEV and WNV infection in adult susceptible mice or i.p. KUNV

in young HeJ mice. Kobiler and co-workers (1989) used i.v. inoculation to deliver

WNV and this route ensured a high virus presence in the blood stream so that the

concurrent SDS treatment would allow virus to enter the brain. However, even when the

virus was delivered direct to the bloodstream, Kobiler and co-workers (1989) estimated

that only 0.1% of the original virus dose entered the brain. Thus, following flavivirus

challenge via i.p. route and concurrent SDS administration as performed in the current

study, it was very likely that less than 0.1% virus was available for entry into the brain

and therefore the amount of virus might have not been sufficient to induce morbidity in

susceptible mice. However, when SDS was inoculated two to three days post infection

to allow viraemia to reach the peak, higher mortality rates and shorter times to death

were observed in susceptible HeJ mice. Thus, the timing of SDS injection is crucial and

its effect on survival only becomes apparent when administration coincides with the

viraemic stage in infected mice.

The inability of SDS to promote a fatal disease in KUNV-infected adult HeJ mice

(Table 4.4) was most probably caused by the lack of tropism for peripheral tissues of

KUNV, as discussed previously, and thus the presence of a breached BBB had no effect

on the infection. However, the age-related susceptibility in peripheral tissues in KUNV-

infected young HeJ mice allowed greater level of virus replication that permitted viral

spread into the brain. The breach of BBB by SDS then resulted in early CNS invasion

of KUNV and therefore early manifestation of fatal encephalitis in young HeJ mice.

Macrophages have the ability to act early in the infection to destroy invading pathogens.

In addition, mouse peritoneal macrophages have also been shown to support flavivirus

growth in vivo and in vitro (Goodman and Koprowski, 1962a; Silvia, 1999), suggesting

a dual role of macrophages in flavivirus infection. In this study, both pathogenic and

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

115

protective roles of macrophages were shown to be dependent on the virus strain as well

as the genetic background of the mice.

Previous work performed in the laboratory demonstrated that thioglycollate given 3

days prior to virus infection increased susceptibility of HeJ mice to i.p. WNV challenge

(Pantelic, 2004). Similarly, we found that thioglycollate treatment increased the

incidence of fatal WNV infection from 60% to 100% as well as reducing the ATD in

susceptible HeJ mice (Table 4.7). This further confirms the possible role of

macrophages in harbouring and disseminating WNV in HeJ mice. Thioglycollate causes

sterile inflammation by attracting macrophages to the peritoneal cavity. Thus, at the

time of virus inoculation (3 days after thioglycollate treatment), permissive macrophage

cells were available at high numbers in the peritoneum and may have facilitated their

contact and infection with WNV. Increased infection of peritoneal macrophages

allowed rapid virus infection and multiplication, resulting in higher release of infectious

virus. In contrast, control mice infected i.p. with WNV only had about 10 times lower

numbers of cells in their peritoneum for virus infection. Indeed, thioglycollate treatment

was previously shown to increase and prolong viraemia in WNV-infected HeJ mice

compared to those that did not receive thioglycollate (Pantelic, 2004). The high and

prolonged viraemia following thioglycollate treatment may have promoted a release of

higher amounts of virus for possible invasion of the CNS and subsequently for the

increased incidence of fatal encephalitis in all HeJ mice.

In addition, it has also been suggested that treatment with thioglycollate impairs

immune responses. Since blood monocytes are known to take up an antigen and to

acquire the DC characteristics for antigen presentation to T cells in the lymph nodes

(Murali-Krishna et al, 1996, Mathur et al, 1983, Itano and Jenkins, 2003),

administration of thioglycollate causes the sequestration of monocytes and potentially

leads to impaired antigen presentation and modification of the host antiviral immune

response. Furthermore, thioglycollate down-regulates MHC class II cell surface

molecule expression on peritoneal cells (Adam and Hamilton, 1984), which may also

contribute to the increased susceptibility of thioglycollate treated mice to WNV

infection.

Interestingly, thioglycollate treatment did not induce fatal encephalitis in susceptible

HeJ mice i.p. infected with KUNV (Section 4.2.3.4.1.). Similarly, it has been

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

116

demonstrated earlier in this laboratory that thioglycollate-treated HeJ mice did not

display enhanced susceptibility to i.p. MVEV infection (Pantelic, 2004). The fact that

thioglycollate treatment only increases the incidence of fatal encephalitis of HeJ mice

during i.p. infection with WNV but not KUNV or MVEV indicates that macrophage

may only play an important role in the pathogenesis and dissemination of WNV in

susceptible HeJ mice. This is probably because WNV has higher tropism for

macrophages than KUNV and MVEV, as will be presented in Chapter 6.

Macrophages contribute to viral clearance by exerting phagocytic activity and

producing cytokines such as IL-1, TNFα and IL-6. Their importance in the host defence

against flavivirus infection has also been demonstrated by the work of Ben-Nathan and

co-workers (1996). Transient depletion of macrophages exacerbates the course of WNV

infection in mice. Mice showed prolonged viraemia and increased incidence of fatal

disease (Ben-Nathan et al, 1996). Similar results were attained in the study described

here. Transient macrophage depletion increased mortality from 60% to 100% and

reduced average survival time of HeJ mice following i.p. challenge with WNV,

although the significance of this result was not possible to estimate due to a small scale

study (Table 4.9). It has been argued that monocytes repopulate the bloodstream 24

hours post treatment, thus clodronate treatment in the current study resulted in the loss

of scavenger cells only at the early stage of infection and macrophages may have been

repopulated in WNV-infected HeJ mice few days after infection. Nevertheless, it was

demonstrated here that even this initial loss of macrophages was sufficient to impair the

host non-specific immunity. In this case, the importance of macrophages for the early

host antiviral immune response to WNV outweighed their involvement in providing

additional sites for extraneural replication of WNV.

It was reported previously that the host immune response works synergistically with the

flavivirus resistance gene for the prevention of fatal disease in resistant mice upon

flavivirus infection (reviewed in Brinton and Perelygin, 2003). This is because, the

flavivirus resistance gene, Flv, only reduces or slows virus replication, while functional

immune response is required to clear the infection (Brinton and Perelygin, 2003).

Several studies have shown that immunosuppression may abrogate the resistance

expressed by RV mice and consequently mice can become susceptible to flavivirus

infection (Camenga et al, 1974; Bhatt and Jacoby, 1976; Jacoby et al, 1980). For

instance, absence of T cells either by T cell depletion or treatment with anti thymocyte

CHAPTER 4

NEUROVERULENCE AND NEUROINVASIVENESS

117

serum rendered RV mice susceptible to i.p. infection with Banzi, causing a high

mortality of these mice (Bhatt and Jacoby, 1976; Jacoby et al, 1980). However, it was

demonstrated in the study here that a depletion of CD4+ T cells in young DUB mice or

both CD4+ and CD8+ T cells in adult DUB mice did not compromise the survival of

WNV infected-DUB resistant mice. The difference observed in this and other studies

can be attributed to the different viruses used. Although brain is the major target organ,

it is a probable that Banzi virus may have a higher tropism for peripheral organs than

WNV, enabling its better peripheral replication in the absence of functional T cells and

consequently facilitating its invasion of the CNS.

The lack of permissive cells and tissues in resistant DUB mice due to the possible

action of the Flvr-like gene was observed in peripheral tissues (Urosevic et al, 2000;

Pantelic et al, 2004; Chapter 5). Indeed, in vitro KUNV, MVEV and WNV infection of

macrophages isolated from resistant DUB mice resulted not only in lower titres but also

in earlier clearance of the viruses compared to cells derived from susceptible HeJ mice

(see Chapter 5). Thus, as shown for the first time in this study, breaching of the BBB,

attracting macrophages to peritoneal cavity by the thioglycollate treatment as well as

depleting macrophage and T cells had no consequence on the survival of resistant DUB

mice following i.p. inoculation of WNV. This suggests that the infectibility of

extraneural cells with this virus may have been compromised by the expression of the

resistance gene, Flvr-like, that prevented a high virus replication at the periphery.

In conclusion, the data presented in this study provide in vivo characterisation of three

closely related flaviviruses in susceptible HeJ and resistant DUB mice, further

confirming that the severity of flavivirus-induced diseases is the result of complex

relations between viral and host factors.. In this study, it was demonstrated that different

flaviviruses express different virulence characteristics in vivo. However, in is not

known whether these three flaviviruses would exhibit similar infectibility in vitro. In

addition, it remains to be confirmed whether macrophages contribute to the

pathogenesis of WNV only and not MVEV and KUV during i.p. inoculation in

susceptible HeJ, as demonstrated by the thioglycollate treatment studies. Thus,

infection of KUNV, MVEV and WNV in cells lines and primary cell cultures including

macrophages would enable studies on in vitro characterisation of these three closely

related flaviviruses as well as on the dual role of macrophages in certain flavivirus

infections, as described in the proceeding chapter.

CHAPTER 5

INFECTION IN CELL CULTURE

118

5.0 CHAPTER 5: CHARACTERISATION OF KUNV, MVEV AND

WNV INFECTIONS IN CELL CULTURE

5.1 INTRODUCTION

As described in the previous chapter, KUNV, MVEV and WNV, although are closely

related flaviviruses, vary greatly in their virulence in vivo. However, very little is

known about their growth characteristics in cell culture. Thus, this part of the study was

intended to characterise the in vitro infectibility of KUNV, MVEV and WNV in parallel

as well as to elucidate the possible role these cells may have in the pathogenesis of

flaviviruses as suggested in the preceding chapter.

A cell culture approach provides a useful model to study growth characteristics and

replication of different viruses in parallel in an isolated and controlled environment. The

first part of the study was aimed at studying the replication of KUNV, MVEV and

WNV in different cell types. The types of cells used were Vero cells, which are

established fibroblast cell line derived from the kidneys of green monkey, primary

peritoneal macrophages derived from susceptible HeJ and resistant DUB mice and

primary bone marrow-derived DCs isolated from susceptible C57BL/6 mice. In

addition to virus growth, pro-inflammatory cytokine production was also investigated in

thioglycollate elicited-macrophages during infection with KUNV, MVEV and WNV.

The pathogenesis and cellular basis of virulence of certain flaviviruses in vivo could be

due to the ability of macrophage to support replication of these flaviviruses. Thus, the

second aim of this study was to further explore the possibility that macrophages are

potential sites for harbouring WNV in the periphery, as suggested by the thioglycollate

experiment performed in the previous chapter (Chapter 4). To achieve this,

thioglycollate elicited-peritoneal cells were infected with WNV in vitro prior to their

administration back into the mice by the i.p. inoculation.

CHAPTER 5

INFECTION IN CELL CULTURE

119

5.2 RESULTS

5.2.1 VIRUS REPLICATION IN CELL CULTURE

In this study, parallel in vitro characterisation of WNV, KUNV and MVEV infection

was performed on Vero cells, primary mouse macrophages and primary murine DCs to

examine whether these cells exhibit similar permissiveness to these three viruses. Vero

cells were included in this study as a reference permissive cell line that was normally

used to propagate flaviviruses in the laboratory.

5.2.1.1 Determination of dose of infection

Infection analysis performed earlier in this laboratory (Lancaster et al, 1998) indicates

that roughly 60% of Vero cells are infected at the initial stage of MVEV infection which

then increased to 100% 1-2 days later. In contrast, no more than 10% and 5% of HeJ

and DUB macrophages, respectively, could get infected upon WNV in vitro infection

(Pantelic, 2004; Silvia, 1999). It was also shown that infection of MVEV at MOI 1 in

primary murine macrophages resulted in low virus replication (Silvia, 1999). Because of

these, MOI 10 (equivalent to 1 x 107 i.u. or 10

6.2/100μL TCID50 units) was selected as a

standard dose of infection for the present study.

5.2.1.2 Virus replication in Vero cells

In the first part of this in vitro study, the infectibility of three flaviviruses, WNV,

KUNV and MVEV was investigated in Vero cells, which served as reference cells for

the virus infection. Vero cells were seeded in 6 well plates at 1 x 106/well and incubated

for 6 hours to allow cell attachment prior to KUNV, MVEV and WNV infection at

MOI 10. To determine the levels of virus replication in culture supernatants collected

from all infected Vero cells at different time points p.i., TCID50 bioassay used.

Vero cells started to die 5 days after being infected and therefore collection of samples

was discontinued beyond this time point. As shown in Figure 5.1, of all three

flaviviruses used in this study, WNV replicated most efficiently in Vero cells, followed

by KUNV and MVEV. The replication of these three flaviviruses increased sharply

from day 1 p.i. and the highest titres were attained on day 2 p.i. However, WNV titres

CHAPTER 5

INFECTION IN CELL CULTURE

120

were a log higher (8.23 ± 0.1 log10 TCID50; significantly different, Student t test, p <

0.05) at this time point compared to KUNV (7.28 ± 0.6 log10 TCID50) and MVEV (7.34

± 0.2 log10 TCID50). Vero cells looked slightly unhealthy 3 days after infection,

particularly in cultures infected with WNV and KUNV. Coincident with the early

cytopathic effect, production of all 3 viruses started to decrease. Two days later, at day 5

p.i., cytopathic effect induced by the virus replication was apparent in cells infected

with the three viruses (Figure 5.4). At this time point (day 5 p.i.), WNV titres were still

the highest (Student t test, p < 0.01) when compared to the other two viruses despite the

replication of all three viruses was slightly reduced by then. The MVEV titres in Vero

cells were consistently the lowest from day 3 p.i. to day 5 p.i.

5.2.1.3 Virus replication in thioglycollate-elicited macrophages

Thioglycollate is a nutrient rich medium that acts as a sterile inflammatory agent when

injected into the peritoneum. Intraperitoneal injection with this compound results in the

migration and accumulation of blood monocytes to the peritoneal cavity. These cells,

which later differentiate at variable rates into macrophages, contribute to a great cellular

diversity in the peritoneum. Thioglycollate-elicited peritoneal cells comprise of

immature, mature and differently activated macrophages, recruited from both the

circulating and marginal tissue pools. Because of this, the cell exudates obtained

following thioglycollate treatment consist of heterogeneous cell populations (van Furth

et al, 1973). In contrast, the peritoneal cavity of un-stimulated mice consists of a more

homogenous population of mature resident macrophages. In addition, thioglycollate

treatment causes a 10-fold increase in cell numbers in the peritoneum, thus enabling

greater yields of isolated cells from the same number of mice when compared to the

non-treated animals (Silvia, 1999).

In order to prepare primary macrophage cultures, peritoneal macrophages were isolated

from susceptible HeJ and resistant DUB mice three days after administration of 1mL

6% thioglycollate. Following isolation, these cells were cultured in vitro in 6-well plates

at 1 x 106 cells/well and they were kept under adherent conditions for 5 days prior to

infection, in order to allow removal of cytokines that may have been generated due to

thioglycollate treatment in vivo and which may cause a resistance to virus infection in

vitro (Pantelic, 2004).

CHAPTER 5

INFECTION IN CELL CULTURE

121

As shown in Figure 5.2A, the peak of KUNV, MVEV and WNV replication in primary

macrophages derived from susceptible HeJ mice was at day 2 p.i., similar to that

observed in Vero cells. Following this, titres of the three viruses in macrophage cultures

started to decline (Figure 5.2). However, titres of KUNV, MVEV and WNV were 2-3

logs lower compared to Vero cells and no virus-induced cytopathic effect was observed

when infected-macrophages were monitored for up to 15 days p.i. (Figure 5.5). WNV

was also demonstrated to be the most efficient virus to infect macrophages while in

contrast, MVEV showed the lowest replication in these cells. Two days after infection,

WNV titres in macrophages were found to be 6.2 ± 0.4 log10 TCID50. This was a log

higher than KUNV titres and 2 logs higher than MVEV production at the same time

point of infection. Virus replication then gradually decreased and interestingly, on day

15 p.i., WNV and KUNV still persisted at low levels in macrophages (2.02 ± 0.7 log10

TCID50 and 0.6 ± 0.1 log10 TCID50, respectively) while infectious MVEV was no longer

detectable at day 13 p.i.

Macrophages derived from resistant DUB mice are known to express the Flv resistance

gene (Pantelic, 2005). Although cells from resistant mice can be infected with

flaviviruses, the rate of infection is much lower than that observed in cells from

susceptible mice (Silvia 1999; Pantelic et al, 2005). In the current experiments,

thioglycollate-elicited macrophages isolated from flavivirus resistant DUB mice were

subjected to parallel infections with these three flaviviruses for the first time. In

agreement with the previous observations, it was demonstrated here that KUNV,

MVEV and WNV replicated 1-2 logs lower in DUB-derived macrophages than in HeJ-

derived macrophages (Figure 5.2). However, while KUNV showed slightly greater

replication than MVEV, WNV still produced the highest virus titres compared to these

two flaviviruses throughout the course of infection. On day 2 p.i., viral production was

4.3 ± 0.3 log10 TCID50, 3.5 ± 0.03 log10 TCID50 and 3.5 ± 0.2 log10 TCID50 for WNV,

KUNV and MVEV, respectively. The production of these viruses then steadily declined.

However, the rate of KUNV, MVEV and WNV decline in resistant macrophages was

much faster than from susceptible macrophages. In addition, similar to susceptible

macrophages, MVEV infection of DUB-derived macrophages showed the most rapid

rate of virus decline in comparison to KUNV and WNV infection. Both MVEV and

KUNV were no longer detectable on day 9 p.i. while WNV was cleared from resistant

macrophages by day 11 p.i.

CHAPTER 5

INFECTION IN CELL CULTURE

122

The findings in this study demonstrated the different ability of KUNV, MVEV and

WNV to replicate in primary murine macrophages. In vitro infection of HeJ or DUB-

derived macrophages showed that WNV and KUNV consistently have higher levels of

replication than MVEV, suggesting that these cells could potentially support in vivo

replication and consequently be involved in the pathogenesis of WNV and KUNV

during i.p. and i.c. infection, respectively.

5.2.1.4 Virus replication in primary mouse dendritic cells

Although infection of the CNS particularly the neurons is the ultimate target of

flavivirus infection and dictates the outcome of infection, DCs represent an early and

very important target cell type for some flaviviruses. In this part of study, parallel

infection of primary DC cell cultures derived from susceptible C57BL/6 mice with three

flaviviruses, KUNV, MVEV and WNV was studied. In the natural route of flavivirus

infection following mosquito bites, DCs in the skin can become infected and carry the

virus to the local lymph nodes for antigen presentation to T cells (King et al, 2003).

This suggests that, in addition to be involved in initiating and activating the host

immune response, DCs could also possibly contribute to the pathogenesis of

flaviviruses by spreading the virus to other organs. In the proceeding study (Chapter 7),

DCs were also found to accumulate in resistant mouse brains when analysed at 9 days

following i.c. challenge with KUNV and MVEV infection. The role of DCs either as

crucial APCs in the CNS or as immunopathogenic agents during the i.c. virus challenge

however remains controversial (McMahon et al, 2006). Thus, this study was performed

to investigate the permissiveness of DCs to KUNV, MVEV and WNV.

The DCs used in this study were derived from the bone marrow of C57BL/6 mice

(kindly provided by Ms Andrea Lee, from the laboratory of Dr MP Degli-Esposti).

These cells were characterised by their dual expression of CD11b+ and CD11c+ cell

surface markers. Previous studies in the laboratory have already shown that C57BL/6

mice were similarly susceptible to i.p. WNV infection and i.c. KUNV inoculation as

susceptible HeJ mice (Pantelic, 2004; Chapter 4).

CHAPTER 5

INFECTION IN CELL CULTURE

123

0

1

2

3

4

5

6

7

8

9

0 1 2 3 4 5Days post infection

Vir

al ti

tres (

log

10 T

CID

50/1

00u

L)

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8 9 10 11Days post infection

Vir

us

titr

es (

log

10 T

CID

50/1

00

uL

)

WNV KUNV MVEV

Figure 5.1. Replication of WNV, KUNV and MVEV in Vero cells

Cells were plated in 6-well culture plates at 1 × 106 cells/well. Cells were infected with

viruses at MOI 10 and cell culture supernatants were collected at different time post

infections. Six samples from 3 separate experiments were assayed at each time points

for viral titres by TCID50 bioassay. Data were shown as average values ± SE.

CHAPTER 5

INFECTION IN CELL CULTURE

124

A

0

1

2

3

4

5

6

7

8

9

0 2 4 6 8 10 12 14

Days post infection

Vir

us t

itre

s (

log

10 T

CID

50/1

00u

L)

B

0

1

2

3

4

5

6

0 2 4 6 8 10 12 14

Days post infection

Vir

us t

itre

s (

log

10 T

CID

50/1

00u

L)

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8 9 10 11Days post infection

Vir

us

titr

es (

log

10 T

CID

50/1

00

uL

)

WNV KUNV MVEV

Figure 5.2. Replication of WNV, KUNV and MVEV in A) thioglycollate-elicited

macrophages from flavivirus susceptible HeJ mice and B) resistant DUB mice.

CHAPTER 5

INFECTION IN CELL CULTURE

125

Cells were plated in 6-well culture plates at 1 × 106 cells/well. Macrophages were

incubated for 5 days before being infected with viruses at MOI 10 and cell culture

supernatants were collected at different time post infections. Vero cells were infected

with the same MOI. Six samples from 3 separate experiments were assayed at each time

points for viral titres by TCID50 bioassay. Data were shown as average values ± SE

CHAPTER 5

INFECTION IN CELL CULTURE

126

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8 9 10 11

Days post infection

Vir

us

titr

es (

log

10 T

CID

50/1

00u

L)

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8 9 10 11Days post infection

Vir

us

titr

es (

log

10 T

CID

50/1

00u

L)

WNV KUNV MVEV

Figure 5.3. Replication of WNV, KUNV and MVEV in C57/BL6 mouse bone

marrow derived dendritic cells.

Cells were plated in 6-well culture plates at 1 x 106 cells/well. DCs were incubated for 5

days before being infected with viruses at MOI 10 and cell culture supernatants were

collected at different time post infections. Three to six samples from 3 separate

experiments were assayed at each time points for viral titres by TCID50 bioassay. Data

were shown as average values ± SE.

CHAPTER 5

INFECTION IN CELL CULTURE

127

A B

C D

Figure 5.4. Cytopathic effect of virus replication in Vero cells.

A) Non-infected Vero cells with normal morphology. Infection with B) WNV C)

KUNV and D) MVEV caused cytopathic effect in Vero cells 5 days after infection.

CHAPTER 5

INFECTION IN CELL CULTURE

128

A B

C D

Figure 5.5. Cytopathic effect of virus replication in macrophage cell cultures.

A) Non-infected macrophage cell cultures. Cell cultures were infected with B) WNV C)

KUNV and D) MVEV on day 15 p.i. The cell cultures were observed daily from day 2

until day 15 p.i. and no cytopathic effect was induced in macrophage cell cultures by

virus replication.

CHAPTER 5

INFECTION IN CELL CULTURE

129

As shown in Figure 5.3, the highest levels of infection of DCs were attained on days 2

and 3 p.i. In addition, WNV consistently replicated to the highest titres throughout the

infection course while KUNV had a lower level of replication than WNV but higher

than MVEV. However, levels of virus production in DCs were significantly lower than

observed in primary macrophages from susceptible HeJ mice at day 2 p.i. (Student t

test, p < 0.004, p < 0.04 and p < 0.004 for WNV, KUNV and MVEV, respectively).

MVEV virus replication sharply declined and by day 7 p.i., infectious virus was no

longer detectable by TCID50 bioassay in DCs from susceptible C57BL/6 mice. Thus,

MVEV was cleared much quicker from DCs derived from susceptible C57BL/6 mice

than from primary macrophages derived from resistant and susceptible mice. While

KUNV persisted at low levels in macrophages from susceptible HeJ mice for 15 days, it

declined much faster in DCs derived from C57BL/6 mice, by day 7 p.i. (Figure 5.3).

Furthermore, WNV was cleared from C57BL/6-derived DCs 4 days earlier than from

HeJ-derived macrophages.

Dendritic cells infected with KUNV, MVEV or WNV did not show any cytopathic

effect when monitored for 15 days after infection (data not shown), indicating a non-

cytopathic infection with flaviviruses, similar to that observed in susceptible

macrophages, although of more transient duration. Combined, these data suggest the

role of DCs in disseminating and establishing flavivirus replication early rather than late

in the infection.

5.2.2 CYTOKINE PRODUCTION IN PRIMARY MOUSE MACROPHAGES

Macrophages are known producers of an array of cytokines in response to various

stimuli. Since WNV, KUNV and MVEV replicated at different levels in macrophages,

it was of interest to study whether these flaviviruses would induce different levels of

cytokines. The production of IFN type I, IFN type II and TNFα were monitored in

infected-primary macrophages derived from susceptible HeJ mice over several days

after infection.

IFN type I was analysed in culture supernatants collected on days 1, 2, 3 and 5 p.i. from

infected macrophages using IFN type I bioassay. IFN type I production was below

detection level on day 1 p.i. after infection with all three viruses (Figure 5.6A). The

maximum production of this cytokine was seen on day 2 p.i. However, WNV induced

CHAPTER 5

INFECTION IN CELL CULTURE

130

the highest production of IFN type I (887.5 ± 72 I.U./mL, Student t test, p < 0.003)

compared with KUNV (312 ± 38 I.U./mL) and MVEV (153 ± 97 I.U./mL). At later time

points after infection (days 3 and 5 p.i.), IFN type I production decreased in all infected

macrophages, which directly correlated with the reduction in virus titres at similar time

points (Figure 5.2).

TNFα expression was measured by a commercial ELISA kit (eBioscience) on days 1, 2,

and 3 p.i. As illustrated in Figure 5.6B, TNFα was below detection level on day 1 p.i.

following infection with KUNV, MVEV and WNV. However, on day 2 p.i., TNFα

expression was induced in macrophages infected with WNV and KUNV with similar

levels of TNFα detected (20.4 ± 0.71 pg/mL and 19.67 ± 6.23 pg/mL, respectively),

while the production of TNFα in MVEV-infected macrophages was still undetectable at

this time point. On day 3 .p.i, WNV infection resulted in the highest level of TNFα

production (45.4 ± 2.4 pg/mL) while the expression of TNFα in KUNV-infected

macrophages remained unchanged (18 ± 3 pg/mL). Macrophages infected with MVEV

showed the induction and low production of TNFα on day 3 p.i. (12 ± 0 pg/mL).

IFNγ which is also known as IFN type II, has both antiviral and immunoregulatory

properties (Boehm et al, 1997). Similar to IFN type I, IFNγ production in infected

macrophages was not observed on day 1 p.i., while its production peaked on day 2

p.i. with 95.6 ± 0 pg/mL, 69.6 ± 13.44 pg/mL and 31.25 ± 0 pg/mL of IFNγ were

detected following infection with WNV, KUNV and MVEV, respectively. On the third

day post infection, IFNγ levels in WNV-infected macrophages drastically reduced to

20.9 ± 5.9 pg/mL (Student t test, p < 0.002). In KUNV-infected macrophages the

average levels of this cytokine dropped to 37.04 ± 9.2 pg/mL while infection of MVEV

resulted in only a slight IFNγ decrease (25.7 ± 38 pg/mL) on day 3 p.i.

Data obtained from these cytokine studies indicated that the production of IFN type I,

TNFα and IFNγ coincided mostly with virus titres. The importance of this finding will

be discussed later.

CHAPTER 5

INFECTION IN CELL CULTURE

131

A

0

200

400

600

800

1000

1 2 3 5

Days post infection

IFN

αβ

(IU

/mL

)

B

0

10

20

30

40

50

1 2 3Days post infection

TN

(p

g/m

L)

WNV KUNV MVEV

0

10

20

30

40

50

1 2 3Days post infection

TN

(p

g/m

L)

WNV KUNV MVEV

Figure 5.6. In vitro cytokine productions by HeJ isolated macrophages following

infection with WNV, KUNV and MVEV.

CHAPTER 5

INFECTION IN CELL CULTURE

132

C

0

20

40

60

80

100

120

1 2 3

Days post infection

IFN

γ (

pg

/mL

)

0

10

20

30

40

50

1 2 3Days post infection

TN

(p

g/m

L)

WNV KUNV MVEV

Thioglycollate-elicited macrophages derived from flavivirus susceptible C3H/HeJ mice

were plated in 6-well culture plates and incubated for 5 days prior to virus infection at

MOI 10. 3 samples of the culture supernatants were collected at different time post

infection and assayed for A) IFNαβ B) TNFα. and C) IFNγ. Presence of IFNαβ was

detected by bioassay while the other two cytokines were examined by ELISA. Three

samples were tested per time point for presence of cytokines. Data shown is average ±

SE.

CHAPTER 5

INFECTION IN CELL CULTURE

133

5.2.3 ADOPTIVE TRANSFER OF VIRUS-INFECTED MACROPHAGES IN MICE

Previous studies (Section 5.2.1) demonstrated that WNV could infect thioglycollate-

elicited macrophages isolated from both susceptible and resistant mice. Additionally,

although macrophages are important in the host non-specific defense, earlier work in

this laboratory (Chapter 4; Pantelic, 2004) indicated that macrophages may also be

involved in the dissemination of virus and the promotion of fatal encephalitis following

WNV infection in susceptible mice. Thioglycollate treatment which recruits

macrophages to the peritoneum increased HeJ susceptibility to i.p. challenge with WNV

(Pantelic, 2004). Thus, further investigation into the pathogenic role of macrophages in

disseminating and spreading virus in resistant mice was conducted. This was performed

by infecting thioglycollate-elicited macrophages in vitro and then transferring these

infected cells into mice. Only WNV was included in this study as this virus was shown

to have higher tropism for macrophages than to KUNV and MVEV.

Thioglycollate-elicited macrophages isolated from susceptible HeJ mice were cultured

under non-adherent conditions. Teflon pots were used for this purpose and the cells

were cultured at a density of 1x 106 cells/mL. When analysed by flow cytometry, about

80-86% of cells showed macrophage morphology (based on the forward scatter and side

scatter analysis) and expressed CD11b+ cell surface markers (data not shown).

Following five days of incubation, macrophages were infected with WNV at MOI 10.

Two days after infection, macrophages cells were centrifuged and washed twice with

PBS to remove any free virus from the media and the cells were resuspended in PBS.

Following this, infected macrophages were injected into mice i.p. and the animals were

monitored for any signs of disease for up to 30 days. The number of macrophages

administered into a mouse was equivalent to the number of peritoneal cells isolated

from one mouse following thioglycollate treatment. As a control, a group of seven

susceptible mice were challenged i.p. with WNV at 2 x 107 i.u./mouse only. As shown

in Table 7.1, four out of seven control HeJ mice infected i.p. with WNV developed fatal

encephalitis. The ATD of the sick mice was 9.5 ± 1.0 days. A negative control

comprising mice that received non-infected macrophages was also included. The control

animals did not develop any illness, indicating that macrophages themselves did not

have any toxic effect on mice. However, all susceptible HeJ mice (8/8) that were given

WNV-infected macrophages developed a fatal disease outcome and were culled.

CHAPTER 5

INFECTION IN CELL CULTURE

134

Table 5.1. Mortality studies following i.p. infection of mice with HeJ peritoneal

macrophages infected in vitro with WNV.

Mouse

strain

Macrophage

transfer

Virus

(WNV) Treatment

Mortality

(no. died/

no. tested)

ATDa

HeJ

(adult)

- + - 57 %

(4/7) 9.5 ± 1.0

+ - - 0 %

(0/5) -

+ + - 100 %

(8/8) 8.5 ± 1.0

DUB

(adult)

- + - 0 %

(0/15) -

+ + - 0 %

(0/10) -

+ + SDS

2 days p.i.

0 %

(0/5) -

DUB

(young) + + -

0 %

(0/5) -

aAverage time to death

Thioglycollate-elicited macrophages isolated from HeJ mice were cultured in non-

adherent status for 5 days. A proportion of macrophage cells were then left uninfected

while the remaining cells were infected with WNV at MOI 10. Two days later, the

infected macrophages were injected i.p. into the mice. Mice were monitored for 30 days

for any signs of diseases. Results were pooled from 2 separate experiments.

CHAPTER 5

INFECTION IN CELL CULTURE

135

Table 5.2. Mortality studies following i.p. infection of mice with DUB peritoneal

macrophages infected in vitro with WNV.

Mouse

strain

Macrophage

transfer

Virus

(WNV) Treatment

Mortality

(no. tested/

no. died)

ATDa

HeJ

- + - 66 %

(2/3) 9.0 ± 0.0

+ - - 80 %

(0/5) -

+ + - 66 %

(2/3) 8.0 ± 0.0

DUB

(3 weeks)

- + - 0 %

(0/5) -

+ + - 0 %

(0/5) -

+ + SDS

2 days p.i.

0 %

(0/5) -

aAverage time to death

Thioglycollate-elicited macrophages isolated from HeJ mice were cultured in non-

adherent status for 5 days. A proportion of macrophage cells were left uninfected while

the remaining cells were infected with WNV at MOI 10. Two days later, the infected

macrophages were injected i.p. into the mice. Mice were monitored for 30 days for any

signs of diseases.

CHAPTER 5

INFECTION IN CELL CULTURE

136

The animals exhibited typical signs of diseases including tremors, hind legs paralysis,

ruffled fur, hunched back and placid tail. Early signs of sickness were observed on days

6 and 7 p.i. and they became more apparent on day 8 p.i. when the majority of mice

were culled. This was in contrast to mice infected i.p. with WNV, where signs of fatal

illness were only observed from day 9 p.i. onwards (Student t test, p > 0.05). In vitro

and in vivo data obtained in this part of the study further supported our findings

presented in the preceding chapter (Chapter 4) on the damaging role of macrophages

during WNV infection. Collectively, it was shown here that macrophages were able to

support high replication of WNV and therefore most probably participated in WNV

dissemination to other peripheral organs and the brain in susceptible HeJ mice.

A similar experiment was also carried out in adult and young resistant DUB mice. As

shown in Table 5.1 and in Chapter 4, i.p. infection of WNV did not cause any mortality

in resistant mice. Similarly, adoptively transferred WNV-infected macrophages also did

not induce fatal encephalitis in adult or young resistant DUB mice. When SDS

(240ng/mouse) was administered into adult DUB mice 2 days after receiving infected

macrophages, mice remained healthy and did not become sick when monitored for 30

days.

The ability of DUB-derived macrophages to be infected with and propagate virus

further was also investigated in a separate small study (Table 7.2). When the WNV-

infected macrophages derived from resistant DUB mice were adoptively transferred to

susceptible HeJ mice, only 2 out of 3 mice succumbed to infection. The mortality rate

was similar to that exhibited by control mice directly infected with WNV although

susceptible HeJ mice receiving WNV-infected macrophages died a day earlier (8 days

p.i.) than the control mice. However, adoptive transfer of infected DUB-macrophages

did not induce death in young DUB mice even when SDS was given two days after the

cell transfer (Table 7.2).

The experiments described above showed that macrophages contributed to i.p. WNV

pathogenesis only in susceptible HeJ mice and not in resistant DUB mice following i.p.

challenge. At present, the inability of macrophage to disseminate flaviviruses to the

CNS of resistant DUB mice is not completely understood although it could be due to the

effect/action of the flavivirus resistance gene, Flvr-like.

CHAPTER 5

INFECTION IN CELL CULTURE

137

5.3 DISCUSSION

Macrophages are important components of the host innate defence mechanisms.

Production of proinflammatory cytokines, nitric oxide and phagocytosis are some of the

antimicrobial actions performed by macrophages to help clear microbial infection

(reviewed in Hendriks et al, 2005). However, macrophages have pleiotropic roles and

they have also been shown to be involved in the pathogenesis of flaviviruses.

Macrophages can serve as sites for flavivirus replication and they may also participate

in ADE of infection, mediated by Fc and complement receptors (Cardosa et al, 1986).

Both mechanisms indicate the pathogenic role of macrophages in flavivirus infection.

This current study was designed to a) study the growth characteristics of KUNV,

MVEV and WNV in Vero cells and two different primary murine cells and b) the role

of macrophages in disseminating flavivirus to the CNS in susceptible HeJ and resistant

DUB mice.

In this study, highly permissive Vero cells were shown to be susceptible to infection

with all three flaviviruses which induced severe CPE several days after infection

(Section 5.2.1.1). In contrast, flavivirus infections did not induce CPE in macrophage or

DCs, similar to observations reported by others (Silvia et al, 2004; Shirato et al, 2006;

Rios et al, 2006). A significant finding of the study is that, of the three flaviviruses

analysed, WNV has the greatest levels of replication for all the cell models studied here.

The higher infectibility of WNV in macrophages particularly those derived from HeJ

mice suggests that these cells could be important for harbouring and dissemination of

WNV in vivo. The macrophage contribution to the severity of WNV infection in HeJ

mice was further confirmed by adoptive transfer of WNV-infected macrophages (to be

discussed later). However, the molecular basis of the ability of WNV to replicate at

higher rates in cell culture than KUNV or MVEV is yet to be elucidated. These three

flaviviruses may carry a number of different virulent determinants that could be

responsible for the variable infection rate observed in vitro as well as different outcomes

of in vivo infection.

The poor infectibility of primary macrophages and DCs with MVEV is interesting

(Figure 5.2 and 5.3). The low replication rate of MVEV in primary cells was observed

despite the same numbers of infectious units of KUNV, MVEV and WNV used for cell

culture infection (1 x 107 i.u. of viruses) and despite the high neuroinvasiveness and

CHAPTER 5

INFECTION IN CELL CULTURE

138

neurovirulence of MVEV in HeJ mice as presented in the preceding chapters (Chapter

4). In contrast to the in vitro findings, i.p. infection with KUNV did not induce any

morbidity in adult susceptible HeJ mice while high death rate in the same mouse strain

was recorded following i.p. challenge with MVEV. Additionally, following i.c.

challenge, a lower amount of MVEV was required to kill 50% of HeJ mice compared to

KUNV. Thus, the finding in this current chapter is intriguing and may indicate that the

mechanisms of MVEV pathogenesis are different from KUNV and may not involve

macrophages and/or DCs.

Macrophages isolated from resistant DUB mice could be infected with flaviviruses as

the genetically determined resistance against flaviviruses does not operate at the level of

virus attachment or entry. Inborn resistance to flaviviruses is reported to function

intracellularly by reducing virus titres and limiting the virus spread (Urosevic and

Shellam, 2002; Brinton, 2001). Because of this, comparable numbers of cells were

shown to be infected in mouse embryo fibroblast (MEF) cultures derived from

susceptible and resistant cultures following challenge with WNV (Brinton et al, 1974).

However, although the titres produced in resistant macrophages usually are lower than

in susceptible macrophages, the difference in titre is usually not as high as observed in

the brain (Silvia et al, 2001; Brinton and Perelygin, 2003). In agreement with this,

KUNV, MVEV and WNV replicated at reduced levels in resistant macrophages cells,

but only 1-2 logs lower compared to susceptible macrophages (Figure 7.1). This is in

contrast to the 3-4 logs brain titre difference observed in susceptible HeJ and resistant

DUB mice during i.c. KUNV and MVEV infection (see Chapter 6). In this project,

2 x 107 i.u. of virus (equals to 10

6.2/100μL TCID50 units) were used for in vitro

infection. However, equal or smaller virus doses were used for the i.c. inoculation in

mice (1.74 x 105 i.u. KUNV and 3.4 x 10

3 i.u. MVEV) which corresponded to

106.7

/100μL TCID50 units and 105/100μL TCID50 units, respectively (see Chapter 6).

Thus, the discrepancy in the initial viral dose in vitro and in vivo is not the cause of the

difference in the extent of resistance/susceptibility observed between in vivo and in vitro

models. In fact, the disparity between these two models of infection can be attributed to

the inability of macrophages derived from susceptible HeJ mice (in vitro) to support as

high levels of flavivirus replication as observed in the brains (in vivo) of susceptible HeJ

mice. So, it appears that the flavivirus replication in primary macrophages is limited to a

single virus life cycle, preventing amplifying step of virus replication as observed in the

CHAPTER 5

INFECTION IN CELL CULTURE

139

brains in vivo and thus keeping the difference in virus titres between susceptible and

resistant macrophages at minimum.

Following natural infection of flaviviruses by mosquito bites, skin DCs are thought to

be the primary cell type to be infected by the virus. In addition, infected DCs transport

the virus to the local draining lymph nodes where the virus is presented to T and B cells

(reviewed in Chambers and Diamond, 2003; Johnston et al, 2000). Using bone-marrow

derived DCs isolated from susceptible C57BL/6 mice, it was demonstrated here that

similar to Vero cells and macrophages, DCs were permissive to KUNV, MVEV and

WNV. Nevertheless, the peritoneal macrophages supported better flavivirus replication

than the bone marrow derived DCs (Figure 5.3). This suggests that the bone-marrow

derived DCs are not the best model for flavivirus infection.

Macrophages can produce an array of cytokines following microbial infection or any

other stimuli. The levels of cytokines; IFNαβ, IFNγ and TNFα in the cell culture

supernatants from infected cells were below detection level on day 1 p.i. (Figure 5.6).

This could due to a lag period where the signalling and activation of cytokine genes

took place. In general, secretion of these cytokines was positively correlated with virus

production. The direct correlation between IFN type I and TNFα and virus titres were

also observed during in vivo i.c. infection of mice with KUNV and MVEV (Chapter 6).

Although both cytokines are known to have direct antiviral properties, the importance

for KUNV, MVE and WNV clearance observed here remains to be investigated. The

use of neutralising antibodies to IFN type I and TNFα, may help to shed more light on

the involvement of these cytokines. Secretion of other cytokines by macrophages such

as IL-1 and IL-6 and their role also remains to be elucidated as well.

Blood-borne macrophages have been implicated in the pathogenesis of Theiler’s murine

encephalitis virus and EAE (Bauer et al, 1995; Brosnan et al, 1981; Rossi et al, 1997).

In Theiler’s virus infection, persistent infection in macrophages has been reported and

depletion of blood-borne macrophages almost completely eliminated viral RNA and

antigen in the CNS (Rossi et al, 1997). The permissiveness of HeJ macrophages to

WNV as well as the ability of the virus to persist in the cells for about 2 weeks suggests

that macrophages also may be involved in the dissemination and pathogenesis of WNV.

Adoptive transfer of in vitro infected thioglycollate-elicited macrophages further

confirmed this theory, as evidenced by the higher mortality exhibited in susceptible HeJ

CHAPTER 5

INFECTION IN CELL CULTURE

140

mice compared to control mice. One hypothesis is that upon transfer of highly infected

HeJ macrophages to the peritoneum of susceptible mice, high titres of infectious virus

would be released in the peritoneal cavity. This would increase the chances of other

susceptible peripheral cells or tissues to be infected. As a consequence, WNV may

replicate at high titres and induce viraemia, enabling this virus to invade the CNS. In

addition, it was shown that WNV infection in macrophages was accompanied by high

levels of TNFα production (Figure 5.6). The toll-like receptor (TLR) 3 dependent

inflammatory responses, particularly TNFα, has been reported to be involved in the

breakdown of the BBB, and subsequently facilitating WNV invasion into the brain

(Wang et al, 2004a). Thus, the production of TNFα by WNV-infected adoptively

transferred macrophages as well as by other peripherally infected cells/tissues would

exacerbate the disease by increasing the permeability of the BBB, enabling the invasion

of infected macrophages into the CNS. Thus, studies shown here further confirmed the

findings of Pantelic (2004) that macrophages may act as ‘Trojan Horses’ to disseminate

flavivirus to the brain.

The increased severity of disease and fatal infection upon adoptive transfer of WNV-

infected HeJ macrophages was only demonstrable in susceptible HeJ mice but not in

adult and young resistant DUB mice. It is possible that although adoptive transfer of

WNV-infected HeJ macrophages in DUB mice may release infectious WNV, the

constitutive expression of the flavivirus resistance gene in extraneural tissues and cells

prevent further replication of WNV in other organs/cells of DUB mice. Consequently,

WNV invasion to the CNS may not occur, resulting in a complete survival of DUB

mice.

In conclusion, findings from this chapter indicated that KUNV, MVEV and WNV could

infect peritoneal macrophages in vitro but replication rate of these viruses was

significantly different. Interestingly, replication levels of these three flaviviruses did not

correlate with their neuroinvasive traits (Chapter 4), suggesting that different pathogenic

mechanisms may be involved following infection with each particular flavivirus tested

in this study. In addition to their protective role, macrophages could also contribute to

the poor outcome of flavivirus infections, although the latter role is greatly regulated by

both virus and host factors. Data in this study further support the previous studies

described in Chapter 4 as well those previously performed in this laboratory (Pantelic,

2004) regarding the possible role played by macrophages in WNV pathogenesis in

CHAPTER 5

INFECTION IN CELL CULTURE

141

susceptible HeJ following i.p. challenge. While macrophages may not play a crucial role

in dissemination of KUNV following i.p. challenge, they may still have an important

role in the pathogenesis of i.c. infection of KUNV.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

142

6.0 CHAPTER 6: ROLE OF VIRAL REPLICATION AND

IMMUNOPATHOLOGY IN DISEASE DEVELOPMENT

FOLLOWING KUNV AND MVEV INTRACEREBRAL

INFECTION

6.1 INTRODUCTION

Laboratory mice provide a suitable small animal model to study flavivirus-induced

encephalitis since they develop similar disease to that observed in humans (reviewed in

Chambers and Diamond, 2003). The recent emergence/reemergence of WNV in several

continents is alarming due to the high incidence of infection and fatal outcome in

humans (Gould and Fikrig, 2004; Gubler, 2002). It is important to understand the

pathogenesis of flavivirus infections and factors that are associated with the fatal disease

outcome since this will allow development of effective antiviral therapies or vaccines.

Host factors including genetic background, age and immune status determine the level

of host resistance to the infection. However, viral factors such as the virus type, dose

and route of infection also modulate the host response and outcome of infection.

Determination of the role for each of these factors is possible only under controlled

experimental conditions (Sabin, 1954).

A model of genetically resistant mice has been created to study the phenomenon of host

genetic resistance to flaviviruses. Although the inborn resistance conferred by Flv

usually protects mice from developing lethal encephalitis by restricting and reducing

viral production in infected cells and organs, flavivirus resistance mice can still be

infected by flaviviruses (reviewed in Urosevic and Shellam, 2002). This resistance is

sometimes incomplete and flavivirus resistant mice may occasionally succumb to i.c.

infection with certain flaviviruses. It was shown in the previous chapter (Chapter 4) that

while MVEV was the most neurovirulent virus in susceptible HeJ mice compared with

KUNV and WNV, the i.c. challenge with this virus in resistant DUB mice did not

induce any apparent disease or morbidity. In contrast, the i.c. infections with KUNV

and WNV caused relatively high mortalities not only in susceptible mice, but also in

resistant mice. This indicates that, in addition to the genetically host resistant trait, viral

factors may also control the severity of disease in flavivirus resistant mice.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

143

The development of lethal disease in flavivirus resistant mice during infection with

certain flaviviruses including KUNV could either be due to the inadequate ability of the

resistance gene (Flv) to restrict virus replication or because of inappropriate response of

the host immune system. Based on findings by other investigators (Sabin, 1952a;

reviewed in Brinton and Perelygin, 2003) and in the previous chapter, we hypothesised

that death in flavivirus resistant DUB mice following KUNV infection was not due to

the ability of KUNV to abrogate the effect of the flavivirus resistance gene, but rather

was caused by an excessive host immune response. In contrast, deaths observed in

susceptible HeJ mice may be associated with several factors including high brain viral

titres and immunopathological diseases. To test this hypothesis, the viral burden in the

brains was examined in both HeJ and DUB mice following i.c. infection with KUNV

and MVEV. Additionally, histopathological analysis of mouse brains following similar

i.c. challenge was conducted to investigate the severity of brain inflammation. Viral

infections in the CNS may induce apoptosis of infected cells and activate brain

microglia/macrophages to produce an array of cytokines. Although the phenomenon of

incomplete protection in flavivirus resistant mice against flaviviruses has been observed

for many years (Sabin 1952a, 1952b), it is not understood yet what pathological events

are associated with the fatal outcome of some flavivirus infections in resistant mice. To

date, no study has been performed to identify factors or mechanisms that determine a

poor outcome of infection with some flaviviruses in resistant mice. Thus, using KUNV,

which kills resistant mice and MVEV, which does not cause any apparent disease in the

same mouse strain, studies aimed at further elucidation of the cause of death or recovery

following i.c. infection with these viruses, particularly in flavivirus resistant DUB mice

were designed and conducted as described in this chapter.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

144

6.2 RESULTS

6.2.1 BRAIN VIRUS TITRES FOLLOWING INTRACEREBRAL INFECTION

6.2.1.2 Analysis of viral titres in mouse brain following KUNV and MVEV infection

Several host and viral factors are involved in shaping the severity of disease following

virus infection. One of these factors is the ability of virus to replicate at high titres in

infected cells. High levels of virus production could disturb the normal function of

target cells, without necessarily causing cell death. Consequently, an infected host may

die, particularly if vital and post-mitotic cells such as neurons are involved.

Development of lethal encephalitis in susceptible mice during flavivirus infections

following the i.c. route of virus inoculation has been reported to be associated with high

viral titres in the brain (Sabin, 1954; Urosevic et al, 1999; Brinton 2001). In contrast,

following i.c. challenge with some flaviviruses which do not induce morbidity in

resistant mice, the peak brain virus titres are usually significantly lower than that

observed in susceptible mice and these viruses are cleared from the brains of resistant

mice (Urosevic et al, 1999). The kinetics of KUNV growth in susceptible and resistant

mice however is not known. Thus, this study was initiated to look at the levels of virus

replication in the brain, especially in flavivirus resistant mice, and the effect of this

replication on mortality or survival of mice following i.c. challenge with KUNV and

MVEV. This would also help to provide further information on whether KUNV could

abrogate the expression of the Flvr-like gene in resistant DUB mice, leading to a high

production of the virus in the brain.

In this study, a viral dose equivalent to 100LD50 as determined in susceptible HeJ mice,

was used as a standard dose for i.c. infection in this study. This dose would kill 100%

susceptible HeJ mice following i.c. infection with either KUNV or MVEV. Since

KUNV and MVEV showed different virulence traits in susceptible HeJ mice, the

calculated 100LD50 doses corresponded to 1.74 x 105 i.u. (10

-1.5 stock virus dilution) of

KUNV and 3.4 x 103

i.u. (10-2.6

stock virus dilution) of MVEV (Table 6.1). Although

the same in vivo lethal doses of KUNV and MVEV comprised of different amounts of

viral infectious units, as determined by the standard virus titration assay in Vero cells, it

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

145

was found here that LD50 doses are more appropriate for the in vivo virulence studies

than the viral infectious units. The equal amount of infectious virus of two different

flaviviruses might induce similar effect on the same cell culture types in vitro but may

possibly generate different results in vivo. Since the objective of this study was to

understand the different pathogenic mechanisms involved during i.c. infection with

KUNV and MVEV, particularly in resistant DUB mice, the same 100LD50 that would

cause similar mortality rate in susceptible HeJ mice was used.

Table 6.1. The KUNV and MVEV doses used for intracerebral infection in mice

Virus

100LD50

Stock virus

dilution

Infectious

units (i.u.)

Titres in Vero cells

(TCID50 units/5uL)

KUNV 10-1.5

1.74 x 105 10

5.4

MVEV 10-2.6

3.4 x 103 10

3.7

100LD50 as determined in susceptible mice was used for i.c. challenge in mice. Data in

the table show the dose expressed as stock dilution, infectious units and TCID50 values

To monitor KUNV and MVEV replication in infected mouse brains, two groups of

flavivirus resistant DUB mice aged between 8-10 weeks were infected i.c. with either

KUNV or MVEV. In parallel, two groups of age-matched susceptible HeJ mice were

also infected with the same viruses and acted as positive controls. Following infection,

brains from three susceptible HeJ mice were harvested separately each day from day 3

p.i. until mice developed fatal disease (day 5 and 6 p.i. for KUNV and MVEV infection,

respectively) and the level of infectious virus in brain homogenates was determined by a

TCID50 bioassay. The kinetics of brain viral replication in resistant DUB mice were

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

146

monitored from day 3 to day 9 p.i. since KUNV-infected resistant DUB mice on

average, succumbed to the infection 9 days after virus challenge (Chapter 4).

As illustrated in Figure 6.1, virus titres in the brains of susceptible mice were quite high

on day 3 p.i. with both viruses. However, MVEV replicated more than a log higher (7.8

± 0.5 log10 TCID50) than KUNV (6.1 ± 0.4 log10 TCID50) in susceptible HeJ mice.

Replication of both viruses increased over time and mice started to show signs of

disease from day 4 p.i onwards. MVEV reached its highest titres (10.0 ± 0.5 log10

TCID50) in the brains of HeJ mice on day 6 p.i. when mice showed typical signs of

severe disease including tremor, hunched back, ruffled fur, placid tail, substantial

weight loss and they had to be euthanised. In contrast, the peak of KUNV replication

was on day 4 p.i. (7.4 ± 0.5 log10 TCID50) and it reached a plateau until mice became

very sick on day 5 p.i. Although the difference in viral titres found in the brains of HeJ

mice 5 days after MVEV and KUNV infection was significant (Student t test, p < 0.05),

these results indicated that high viral production was associated with the deaths seen in

both KUNV and MVEV-infected susceptible HeJ mice.

In flavivirus resistant DUB mice, the Flvr-like gene acts to reduce viral replication early

in the infection (Urosevic and Shellam, 2002). In support of this, KUNV and MVEV

replicated at significantly lower levels, 3-4 logs lower in the brains of resistant DUB

mice than in the brains of susceptible HeJ mice throughout the course of infection, when

monitored from day 3 to day 9 p.i. (Figure 6.1). Both KUNV and MVEV reached their

peak of replication in the brains of DUB mice on day 5 p.i. Similar to that observed in

susceptible HeJ mice, MVEV titres (5.3 ± 0.5 log10 TCID50) were also higher than

KUNV (4.7 ± 0.1 log10 TCID50) in the brains of resistant DUB mice. After day 5 p.i, the

replication of both viruses declined. Interestingly, while production of infectious MVEV

rapidly decreased and was undetectable by TCID50 bioassay 9 days after infection,

KUNV titres declined at a slower rate and virus was still present in the brains of most of

the infected resistant DUB mice, although at a very low titres (Figure 6.1). The

difference in the rate of virus replication between MVEV and KUNV was significant

on day 8 p.i. (Student t test, p < 0.05). In eight resistant DUB mice that succumbed to

KUNV infection, only one mouse had undetectable virus in the brain while the

remaining seven animals had brain viral tires between 0.5 log10 TCID50/100μL to 3.6

log10 TCID50/100μL.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

147

It may be concluded from this study that susceptible HeJ mice suffered fatal

encephalitis following i.c. challenge with KUNV and MVEV as they were unable to

limit brain virus production and spread, which may have caused improper functioning

and damage to infected neurons. Despite incomplete clearance of the virus in the brains

of KUNV-infected resistant DUB mice at the time of death, this study demonstrated that

Flvr-like phenotype was not abrogated and virus production was significantly restricted

in moribund resistant DUB mice in comparison to sick susceptible HeJ mice following

KUNV infection.

6.2.1.2 Analysis of viral titres in peripheral organs following KUNV and MVEV

infection

Since abundant virus replication was not observed in the brains of resistant DUB mice

succumbing to KUNV infection as shown in the previous section, a possibility still

existed that these mice succumbed to profound extraneural virus replication. When virus

is inoculated i.c., it is believed that a proportion of virus leaks to the periphery and it

may infect the peripheral organs. In order to clarify the possibility of profound virus

replication in peripheral organs as a possible cause of death, the presence of infectious

virus was tested by TCID50 bioassay in the spleen, liver and kidney of HeJ mice at the

time of death following KUNV and MVEV i.c. challenge. As shown in Table 6.1, no

infectious virus was found in the kidneys of mice infected with either virus. However,

some spleens and livers from KUNV and MVEV-infected HeJ mice had low amounts of

infectious virus. Similar analysis was also performed in the spleen, liver, kidney,

adrenal gland and pancreas tissue harvested from infected resistant DUB mice on 3, 5, 7

and 9 days p.i. However, no infectious virus was detected in any of the organs tested

(data not shown).

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

148

0

2

4

6

8

10

12

3 4 5 6 7 8 9

Days post infection

Bra

in v

iral

titr

es (

Lo

g10T

CID

50/0

.01g

)

MVEV-HeJ MVEV-DUB

KUNV-HeJ KUNV-DUB

*

*

Figure 6.1. Kinetics of viral replication in mouse brains infected with KUNV and

MVEV.

Separate brain homogenates from three to five mice (except for DUB infected with

KUNV and MVEV at day 9 p.i where six to eight mice were used) were analysed

independently to determine viral titres for each time points. The assays sensitivity limit

is 1 log10 TCID50/0.01g. *Significant difference in the average brain virus titres during

KUNV versus MVEV infection were observed on day 5 p.i. and day 8 p.i. in HeJ and

DUB mice, respectively (Student t test, p < 0.05). Brain titres in KUNV-infected mice

on day 9 p.i. and MVEV-infected mice on day 8 p.i. were arbitrary values.

The threshold for this assay is 2.0 log10 TCID50 units for accurate detection of virus

titres, which corresponds to 0.7 × 102 i.u./100μL. As virus was also detectable below the

threshold, these values were expressed by arbitrary values a) 1.0 log10 TCID50 units for

virus titres detected between 0.74 × 102 and 0.35 × 10

2 i.u. b) 0.5 log10 TCID50 units for

virus titres detected between 0.35 × 102 and 0.10 × 10

2 i.u. The virus was not detectable

below 0.10 × 102 i.u./100μL by this assay.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

149

6.2.2 BRAIN HISTOPATHOLOGICAL AND INFLAMMATION ANALYSIS

6.2.2.1 Brain architecture and inflammation in KUNV and MVEV infection

The study described above indicated that the death of susceptible mice was associated

with high viral titres following i.c. KUNV and MVEV infection. In contrast, resistant

DUB mice that succumbed to i.c. KUNV had low brain viral titres. Substantial evidence

is available indicating that the flavivirus-infected susceptible host may also die due to

an excessive immune response causing tissue damage, in addition to a direct virus-

induced cytopathic effect on the cells (Wang et al, 2003b; reviewed in Chambers and

Diamond, 2003). In order to address the possibility that other factors besides high brain

viral burden may contribute to the death of infected HeJ mice, the extent of brain

inflammation and tissue damage, as well as microglia/macrophages accumulation and

activation in infected susceptible HeJ mice following KUNV or MVEV i.c. challenge

were monitored. Similar histopathological analysis was also performed in infected

resistant DUB mice. This would determine the role of host immune response during

KUNV and MVEV i.c. challenge in resistant DUB mice and test the current hypothesis

that lethal infection observed in KUNV-infected DUB mice was due to excessive brain

inflammation.

6.2.2.2 Brain tissue architecture and leucocytic infiltration in the brains of infected mice

In order to study differences in the brain tissue architecture as well as the accumulation

of immune cells in the brains of mice infected with different flaviviruses, brains from

infected HeJ and DUB mice were harvested at selected time points after infection (at

least 3 brains per time point) and paraffin-embedded brain tissue sections were

prepared. To allow histopathological analysis, brain tissue sections were stained with

haematoxylin and eosin (HE). In susceptible HeJ mice, pathological changes in the

infected brains were examined at day 3 p.i. and at the time of death; day 5 p.i. for

KUNV and day 6 p.i. for MVEV. In resistant mice, similar analysis was performed in

the brains harvested at days 3, 5 and 9 p.i. after KUNV and MVEV i.c. challenge. The

brains from three uninfected HeJ mice were also included in this study as negative

controls.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

150

Table 6.2 Analysis of viral titres in peripheral organs of KUNV or MVEV-infected

HeJ mice at the time of death.

Virus Virus titres ( log10 TCID50/0.01g)a

Spleen Liver kidney

KUNV

0.5

0

0

0

2.5

2.4

0

0

0

MVEV

0

0

2.0

0

0.5

0

0

0

0

aEach spleen and kidney sample represents a pool of 3 mice per sample. Virus titres

obtained from livers were analysed independently (1 liver per mouse per sample).

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

151

Uninfected control mice showed neither abnormalities in the neuron integrity nor

abnormal immune cells infiltration (Figure 6.2.). Three days after infection with both

viruses, a mild brain inflammation was observed in HeJ mice (data not shown).

However, at the time when HeJ mice succumbed to i.c. KUNV or MVEV infection,

classical evidence of acute encephalitis characterised by perivascular lymphocytic

infiltration, perivascular cuffing, parenchymal infiltration and severe vascular

congestion was observed in the brain tissue sections (Figure 6.3.A, B, C and D.).

Mononuclear cells were the predominant immune cells observed in these mice (Prof. J.

Papadimitriou, pers. comm.). Dilated blood vessels and capillaries with leucocytes

accumulated around them could be seen in different regions throughout the brain.

Additionally, oedema was also observed in some parts of the brain including the

thalamus and meninges. Interestingly, cytoplasmic rarefaction with rounding of

neurons was a prominent feature in the thalamus of HeJ mice following KUNV but not

MVEV infection (Figure 6.3.B). It is not known whether the neuronal dysfunction in the

thalamus contributed to the early death of KUNV-infected HeJ mice. However, despite

the high viral load that was associated with the death of susceptible HeJ mice, only a

small number of neurons showed characteristics of apoptotic or necrotic deaths. While

physical damage to the neurons was not the major feature of late infection, death of

susceptible mice could be attributed to a) a high virus replication in the neurons and b)

extensive brain tissue inflammation, both of which may have affected the normal

functioning of the brain cells. Interestingly, while both KUNV and MVEV induced

severe tissue inflammation in the brains of infected susceptible mice, the severity of the

inflammatory response appeared more pronounced in mouse brains infected with

MVEV than KUNV (Figure 6.3, Table 6.2).

A very mild brain inflammatory response was observed on day 3 p.i. in resistant DUB

mice following i.c. infection with both KUNV and MVEV (data not shown). However,

on day 5 p.i., the brain inflammation in the brains of resistant DUB mice was

mild/medium compared to that seen in the brains of dying susceptible HeJ mice on days

5 or 6 p.i. (Figure 6.4). Fewer dilated blood vessels and leucocytic infiltration were

observed in resistant mice than in susceptible mice at this time point. This coincided

with the low virus titres as shown in Figure 6.1. The extent of brain inflammation then

increased on day 9 p.i. with both virus infections, as more recruitment and extravasation

of immunoinflammatory cells in the parenchyma and meninges were detected (Figure

6.5, Table 6.2). Similar to that demonstrated for susceptible HeJ mice, analysis on

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

152

brains sections taken from infected resistant DUB mice (3-5 mice per virus) suggested

that MVEV also induced a slightly stronger tissue inflammation than KUNV in resistant

DUB mice. This finding was quite intriguing since MVEV did not induce any disease in

resistant DUB mice despite the slightly stronger brain tissues inflammation and greater

virus replication. The difference in the extent of brain tissue inflammation between

KUNV and MVEV in resistant DUB mice was more apparent on day 9 p.i (Figure 6.5).

From this study, it was shown that the extent of brain inflammation was associated with

virus titres but interestingly, not necessarily with the morbidity in mice. MVEV

replicated to higher titres in the mouse brains than KUNV and therefore possibly

induced stronger immune response. It may be concluded that the death of susceptible

HeJ mice infected with either KUNV or MVEV following i.c. challenge was associated

with an excessive host immune response, in addition to the uncontrolled virus growth as

discussed in Section 6.2.1. In contrast, stronger immune response was linked to MVEV

clearance and recovery of infected resistant DUB mice. Thus, this finding rejects the

hypothesis that overly stimulated host immune response (in general, without focusing

on specific inflammatory cells) was the cause of death of KUNV-infected DUB mice.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

153

Figure 6.2. Brain tissue section from uninfected mouse.

Uninfected HeJ mice showed normal and healthy neurons with no abnormalities in the

meninges and brain parenchyma, indicated by the absence of haemorrhage and lack of

immune cells infiltration. Picture shown above is representative of a number of tissue

sections obtained from 3 healthy mice. Magnification x200.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

154

A

B

C

D

Figure 6.3. Brain tissue inflammation in susceptible HeJ mice infected with KUNV

(A and B) or MVEV (C and D) at the time of death.

Mouse brains were harvested from sick HeJ mice on day 5 and day 6 p.i. following

KUNV and MVEV infection, respectively (time of death after challenge with each

virus). Figures shown above are sagittal brain tissue sections stained with haematoxylin

and eosin. A small arrow indicates dilated blood vessels and leucocytic infiltration. A

large arrow indicates meningeal leucocytic infiltration while an arrow without tail

shows swollen neurons. Magnification x200.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

155

A

B

C

D

Figure 6.4. Brain tissue inflammation on day 5 p.i. in DUB mice infected with

KUNV (A and B) or MVEV (C and D).

Figures shown above are sagittal brain tissue sections stained with haematoxylin and

eosin, taken from at least 3 different mice. A small arrow indicates dilated blood vessels

and leucocytic infiltration. A large arrow indicates meningeal leucocytic infiltration.

Magnification x200.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

156

A

B

C

D

Figure 6.5. Brain tissue inflammation on day 9 p.i. following i.c. KUNV (A and B)

and MVEV (C and D) infection in DUB mice.

Figures shown are representative of sagittal mouse brain sections stained with

hematoxylin and eosin, taken from at least 3 different mice. A small arrow indicates

leucocytic infiltration and dilated blood vessel, while a large arrow indicates leucocytic

infiltration at leptomeninges. Magnification x200.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

157

6.2.2.3 Analysis of accumulation and activation of microglia/macrophages in the brains

of virus-infected mice

Microglia are cells of the innate immune system in the brain and comprise 20% of total

glial cell population (Raivich and Banati, 2004; Gehrmann, 1996; Streit et al, 2004).

Microglia are thought to be of haematopoetic origin and share many characteristics with

macrophages (Perry and Gordon, 1988). In a normal uninfected brain, microglia reside

in the parenchyma while brain macrophages are normally found in the perivascular

space and meninges (Raivich and Banati, 2004). Following CNS insults, microglia and

brain macrophages are among the first cell types to respond and get activated. Activated

microglia/macrophages can produce an array of cytokines, NO and other inflammatory

stimuli or molecules in a response to intruding pathogens (Streit, 2002). Other host

immune cells respond to these signals and as a result, peripheral leucocytes including

blood borne monocytes which later can differentiate into macrophages, are recruited to

the brain. However, overproduction of the inflammatory stimuli is toxic to host cells

and in many CNS-related diseases such as in experimental allergic encephalomyelitis

and multiple sclerosis (Raivich and Banati, 2004), activated microglia/macrophages

have been implicated in the disease development and severity. Although studies on

brain inflammation (above) in resistant DUB mice indicated that the mortality observed

following KUNV i.c. challenge was not caused by excessive immune response, this

does not exclude the contribution of particular inflammatory cell types to the death of

mice. Thus, immunohistochemical analysis of activated microglia/macrophages was

important for obtaining evidence regarding the role of these cells in the pathogenesis of

both KUNV and MVEV.

A study of activated microglia/macrophages using tomato lectin was performed on

paraffin-embedded brain tissue sections prepared as described in Section 3.6.1.3. The

immunohistochemical staining using tomato lectin could not distinguish between

activated microglia and macrophages. Therefore, cells that stained positive in this study

were grouped together as activated microglia/macrophages. As shown in Figure 6.6 and

6.7, there were differences in morphology and numbers of microglia/macrophages

between early and late infections with both viruses. Almost no activated

microglia/macrophages could be detected in the brains of both susceptible and resistant

mice on day 3 p.i (data not shown). In contrast, brains harvested from susceptible mice

at the time of death showed an abundance of activated microglia with no significant

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

158

difference observed between KUNV and MVEV infections. The numbers of

microglia/macrophages were 92 ± 6 cells/100µm2 and 85 ± 7 cells/100µm

2 in KUNV

and MVEV-infected HeJ, respectively, at the time of death (Table 6.2, Student t test, p

> 0.05). Microglia/macrophage numbers were obtained by counting positively stained

cells on different sections of the brain including cortex, thalamus, hypothalamus, mid

brain, pons, hippocampus and medulla oblongata. The numbers presented in Table 6.2

represent the average values per total brain. In resistant mice, the activated microglia

could be observed for the first time on day 5 p.i. throughout the brains following

challenge with both viruses. The numbers of activated microglia in brains of resistant

mice 5 days after infection with KUNV were found to be double (79 ± 6 cells/100µm2)

the numbers of activated microglia observed 5 days after MVEV infection (40 ± 6

cells/100µm2) in the same mouse strain. On day 9 p.i, the number of microglia increased

in both virus infections (Figure 6.7F and G, Table 6.2) and at this stage unexpectedly,

there were significantly more activated microglia/macrophages (Student t test, p < 0.03)

present in mouse brains infected with MVEV (105 ± 7 cells/100µm2) than KUNV (85 ±

6 cells/100µm2). Additionally, the staining intensity and hairy structures on the cells

also increased, indicating that microglia seen on day 9 p.i. were more activated than

those observed earlier, on day 5 p.i. (Figure 6.7).

The finding in this set of experiment regarding the smaller numbers of activated

microglia detected in mouse brains i.c. infected with KUNV than in those i.c. infected

with MVEV later in the infection, suggests that the activated microglia may not have a

determining role in the fatal outcome of disease following KUNV challenge in resistant

DUB mice.

6.2.2.4 Contribution of apoptosis to fatal outcome of infection

Apoptosis is a programmed cell death by which unwanted cells are eliminated either

during development or infection. It is characterised by rounding of cells, chromatin

condensation and fragmentation of cellular DNA and blebbing of the plasma membrane

that finally leads to a breakup of cell contents into the membrane bound apoptotic

bodies (Levine et al, 2002). Apoptosis is a mechanism that may be effective to prevent

virus maturation and spreading, but sometimes it can be costly to the host, especially

when the post-mitotic cells such as neurons are involved.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

159

Since virus infection can directly induce apoptosis, and neuronal cell death has been

previously implicated in infections with DENV and WNV (reviewed in Chambers and

Diamond, 2003), this study was undertaken to examine whether KUNV and MVEV

infections in both susceptible HeJ and resistant DUB mice could induce excessive

apoptosis. Furthermore, brain tissue inflammation analysis (section 6.2.2.1) suggested

that apoptosis is not abundant during i.c. flavivirus infection. Thus, this study would

provide further confirmation on this event. The TUNEL assay used in this study detects

fragmentation of DNA, which corresponds to the late stage of apoptosis. Analysis was

performed on susceptible HeJ mice at the time of death (days 5 and 6 p.i.) and on days

5, 7, and 9 p.i on resistant mice. As negative controls, brain tissue sections from

uninfected mice were included in this assay. Additionally, separate tissue sections from

uninfected mice were treated with DNase1 to induce DNA fragmentation and acted as

positive controls. In susceptible mice, there was no difference between the numbers of

apoptotic cells seen following infection with either KUNV or MVEV (Figure 6.8A and

6.8B). Apoptosis was a sporadic event in both infections, with about two to six dead

cells detected in the hippocampus, thalamus as well as in the cortex. The same regions

in the brains of resistant DUB mice were also shown to have some apoptotic cells

following KUNV and MVEV infections on day 9 p.i. (Figure 6.8C and 6.8D). However,

very few cells were detected, approximately one to four dead cells per the similar

regions as in susceptible HeJ mice. There was no apoptotic cells observed on days 5 and

7 p.i. following infection of KUNV and MVEV in DUB mice (data not shown). Cells

that tested positive for apoptosis had morphological characteristics of neurons and

mononuclear cells.

Since only small numbers of dead cells were detected to be positive by the TUNEL

assay, it can be concluded that apoptosis is not the major pathogenic mechanism

associated with the death of either susceptible HeJ or resistant DUB mice during i.c.

KUNV and MVEV challenge.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

160

A

B

C

Figure 6.6. Detection of activated microglia/macrophages in the brains of

susceptible mice following i.c. KUNV and MVEV infection.

(A) Uninfected brain (B) HeJ with KUNV at day 5 p.i. (C) HeJ with MVEV at day 6 p.i.

(D) DUB with KUNV at day 5 p.i. (E) DUB with MVEV at day 5 p.i. (F) DUB with

KUNV at day 9 p.i. (G) DUB with MVEV at day 9 p.i. Figures shown are

representative of mouse brain tissue sections taken from 3 different mice. Arrows

indicate activated microglia/macrophages. Magnification x200.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

161

D

E

F

G

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

162

Table 6.3. Summary of viral titres, histopathology and microglia analysis in

susceptible and resistant mice following i.c. infection with KUNV and MVEV.

Virus Mouse

strain

Days post

infection

bViral titres

(Log10

TCID50/0.01g

brain

bLeucocytic

infiltration

and

inflammation

bMicroglia

count

(/100um2)

KUNV

HeJ a5 7.2 0.2 4 92 6

DUB 5 3.8 0.4 2 79 6

DUB a9 c

1.3 0.6 ¾ 85 6

MVEV

HeJ a6 10.1 0.1 5 85 7

DUB 5 4.4 0.2 3 40 6

DUB 9 0 4/5 105 7

aMice were showing terminal signs of diseases.

b3-5 mice were used for each time points for viral titres, histology and microglia studies

cValue obtained from arbitrary values (refer to section 3.7.6 for method of calculation)

Tissue inflammation and leucocytic infiltration

5 - very severe

4 - severe

3 - slightly severe

2 - medium

1 - mild

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

163

A

B

Figure 6.7. Analysis of apoptosis in brains of susceptible and resistant mice

following i.c. KUNV and MVEV infection.

Very few brain cells were apoptotic as shown by TUNEL in A) HeJ infected with

KUNV day 5 p.i. B) HeJ infected with MVEV day 6 p.i. C) DUB with KUNV day 9

p.i. and D) DUB with MVE day 9 p.i. Arrows indicate apoptotic cells. Magnification

x400.

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

164

C

D

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

165

6.3 DISCUSSION

Resistance against flaviviruses conferred by the Flv gene has been shown to be

incomplete against several flaviviruses (Sabin, 1954; Shueb et al, 2005). Some strains

of flaviviruses can still inflict fatal disease in resistant mice, particularly following i.c.

challenge. In the 1950s, Sabin demonstrated that the French neurotropic strain of YFV

induced some mortality in resistant RV mice after i.c. infection (Sabin, 1954).

Additionally, MVE OR155, MVE OR156, Banzi virus and WNV E101 were also

shown to have the ability to ‘evade’ the resistance expressed in flavivirus resistant mice

and cause lethal infection during i.c. challenge (Sangster et al, 1998; Jacoby and Bhatt,

1976; Hanson et al, 1969). In the current study, the pathogenesis of neurovirulent

flavivirus in resistant DUB mice was studied for the first time.

While the morbidity studies following KUNV, MVEV and WNV challenge in

susceptible HeJ and resistant DUB mice were described in Chapter 4, in this chapter,

studies on histopathological changes and brain tissue inflammation associated with the

deaths of susceptible and resistant mice during i.c. flavivirus infection are described.

This is the first study initiated to shed further understanding on why certain flaviviruses

(such as KUNV) cause fatal infection in resistant DUB mice while others (such as

MVEV) are avirulent in this mouse strain despite their higher virulence in susceptible

HeJ mice. Studies using susceptible HeJ mice were also performed in parallel to look at

whether risk factors involved in the pathogenesis of KUNV and MVEV in resistant

mice were similar to that observed in susceptible mice.

The pathogenic mechanisms associated with the virus-induced neurological diseases are

complex. However, in general, a disease severity is either caused by direct virus

replication or by accumulation and activation of resident or recruited inflammatory cells

in response to the infection (Anderson, 2001; Brehm et al, 2004). In this study, the

morbidity and brain tissue inflammation of KUNV and MVEV-infected susceptible HeJ

mice was caused by several factors including high levels of viral replication in the brain.

Considerably higher viral titres were observed in susceptible HeJ mice compared to

resistant DUB mice (Section 6.2.1.1). Neurons are the primary site of flavivirus

replication in the brain (reviewed in Diamond and Engle, 2003). Hase and co-workers

(1990a, 1990b) demonstrated that in JEV infection, virus grows exclusively in the

neuronal secretory system including RER and Golgi apparatus and as a result, the

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

166

cytoplasmic membranous organelles of infected neurons are destroyed. Consequently,

neuronal dysfunction of a large number of infected neurons rather than cell

lysis/apoptosis led to a fatal disease outcome (Hase et al, 1990b, McMinn et al, 1996;

Johnson et al, 1985). In fact, the role of apoptosis in CNS cells in flavivirus

pathogenesis is controversial since challenges with different flaviviruses have not

provided conclusive observations. Neuroadapted DENV has been shown to induce

apoptosis in young mice (Despres et al, 1998). WNV infection in adult hamsters also

generated similar results (Xiao et al, 2001) thus prompting suggestion that

neurovirulent flavivirus is associated with the extent of neuronal cell death it can induce

(Chambers and Diamond, 2003). In contrast, although high mortality occurred in Swiss

mice challenged with MVEV strain BH3479, the occurrence of apoptosis was very low,

only seen in less than 1 per 1000 infected neurons (Andrews et al, 1999). Results

presented in the current study also demonstrated that neurovirulent MVEV and KUNV

did not induce abundant apoptosis in either susceptible HeJ or resistant DUB mice

(Section 6.2.2.3). Thus, no association could be established between apoptosis and

mortality observed in KUNV and MVEV-infected susceptible HeJ mice or KUNV-

infected resistant DUB mice. This disparity may be attributed to the different mouse and

virus models, route of inoculation and virus doses used here and by other investigators.

Thus, KUNV and MVEV-infected susceptible HeJ mice died probably due to a high

viral replication within the neurons and widespread infection within the brain, resulting

in an extensive neuronal dysfunction, as suggested by Hase and co-workers during JEV

infection (1990a, 1990b). In addition to a small number of apoptotic neurons, some of

the apoptotic cells detected morphologically resembled mononuclear cells. This could

be because in the CNS, when inflammatory cells, particularly T cells, do not come in

contact with an antigen and are no longer needed, they may undergo apoptosis (Dorries,

2001).

Interestingly, KUNV induced mortality a day earlier and at lower titres than MVEV in

susceptible HeJ mice. In fact, MVEV replicated to 3 logs higher in the brains of

susceptible mice before they succumbed to the disease (Section 6.2.1.1). This is

intriguing since infection with KUNV and MVEV produced similar viral titres in Vero

cells (see Chapter 5). One possibility is that KUNV may have slightly different

neurotropism than MVEV, targeting vital brain regions which then may induce disease

earlier. Additionally, neurons from various parts of the brain are known to have a

heterogenous response to IFNγ and its antiviral action, resulting in a failure to restrict

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

167

virus replication at the level of individual neurons and in localised areas of the brain

(Binder and Griffin, 2001). This possibility may be supported by the cytoplasmic

rarefaction and swollen neurons observed in the thalamus of HeJ mouse brains

challenged with KUNV (Figure 6.3). In contrast, neurons with these features were

randomly seen throughout the brain and they were not predominant in the thalamus of

MVEV-infected mouse brains. The thalamus has several functions including regulating

the states of sleep and wakefulness (Steriade and Llines, 1988). Damage to the thalamus

can lead to permanent coma. Previous studies performed in the laboratory have shown

that following i.c. MVEV infection, viral RNA was found in the thalamus, cerebral

cortex, hypothalamus, olfactory tuberculum and bulbs, corpus striatum, medulla

oblongata and hippocampus in susceptible mouse brains (Silvia et al, 2004). However,

the spread and neurotropism of KUNV has not been investigated yet. Thus, it would be

interesting to compare the viral spread of KUNV and MVEV in the brains of HeJ mice.

Such a study would assist in determining whether the involvement of different infected

brain regions during KUNV and MVEV infection contributes to the different outcomes

of infection, as observed in both susceptible and resistant mice.

A possible explanation for the different outcomes of i.c. KUNV versus i.c. MVEV

infection in resistant DUB mice is that KUNV, unlike MVEV, has the ability to

abrogate the Flvr-like-controlled resistance, resulting in a high KUNV replication in the

brains of dying resistant DUB mice. As shown in Figure 6.1, MVEV was rapidly

cleared from the resistant mouse brains and was no longer detectable on day 9 p.i. by

TCID50 bioassay. Similar clearance of infectious MVEV from the brains of infected

resistant mice has also been demonstrated in this laboratory (Urosevic et al, 1999).

Similarly, KUNV-infected resistant DUB mice displayed restricted virus replication in

the brains although they became very sick on day 9 p.i., indicating that the flavivirus

resistance gene, Flvr-like, was fully operational and was not abrogated in these mice

(Figure 6.1). However, the death of KUNV-infected resistant DUB mice was associated

with low KUNV titres in the brain. Sabin (1954) reported that a proportion of resistant

RV mice died after i.c. infection with French neurotropic strain of YFV and dying mice

had similar levels of low virus titres compared to that observed in surviving infected RV

mice. In contrast, brain viral titres found in dying RV mice challenged i.c. with WNV

E101 showed an increase compared with infected resistant RV mice that did not exhibit

any signs of disease, although the viral titres were still much lower than that found in

susceptible HeJ mice and the death of resistant mice occurred three to four days later

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

168

than in susceptible mice (Brinton, 1986; Brinton and Perelygin, 2003), suggesting a

partial abrogation of the flavivirus resistance gene in WNV E101-infected resistant RV

mice. Similar high levels of virus replication in the brains of moribund resistant mice

were also reported following Banzi infection in RV mice (Bhatt et al, 1981). In this

laboratory, fatal infection of resistant mice by MVEV OR155 and MVEV OR 156

which are not highly virulent in susceptible mice has been demonstrated (Urosevic et al,

1999; Sangster et al, 1998). MVEV OR 156 is a very unusual strain of MVEV and

exhibits a low i.c./i.p. LD50 ratio in susceptible mice as well as a low infectious virus in

cell culture (Urosevic et al, 1999). Additionally, this virus has 9% divergence in the 5’

portion of the genome from the MVEV strain that was used in the current study

(Poidinger et al, 1996). The differential outcomes of infection by various closely

related flaviviruses as well as the inconsistent brain viral titres (low or high) in resistant

mice that died from certain flavivirus infections suggest two possible scenarios. These

are either non-abrogation (low virus titres as observed in this study) or partial

abrogation of the flavivirus resistance gene effect (high brain viral burden as reported

by Brinton (1986)) in dying resistant mice. In this study, the cause of death of resistant

mice that displayed low brain virus titres was investigated

The inability of KUNV to be cleared from the brains of infected DUB mice is quite

unusual (Figure 6.1). The reason for this incomplete virus clearance is not known. Apart

from the possibility that KUNV may replicate exclusively or to greater levels in neurons

of certain brain regions as mentioned above, it is also possible that KUNV may infect

other CNS cells including brain microglia and macrophages, both of which may lead to

the evasion from host immune surveillance and virus persistence at low titres. Although

flaviviruses have been shown to replicate primarily in neurons in vivo, in vitro infection

demonstrated that a variety of other CNS cells such as astrocytes and oligodendrocytes

can also be infected (reviewed in Chambers and Diamond, 2003; Abraham and

Manjutah, 2006). Cheeran and co-workers (2005) demonstrated that human microglia

could not be infected in vitro, while a persistent JEV infection has been reported in

monocytes (Yang et al, 2004). However, the possibility of brain microglia and

monocytes-derived macrophages as another site of virus infection could not be excluded

for KUNV. A future study whereby brain mononuclear cells will be separated according

to cell surface expression (CD4+, CD8+, and CD11b+) by flow cytometry and tested for

the presence of viral RNA by RT-PCR may yield more information about the

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

169

involvement of microglia/macrophages or other inflammatory cells in the pathogenesis

of flaviviruses.

Unexpectedly, a greater level of brain inflammation was observed following i.c. MVEV

than i.c. KUNV infection. However, higher levels of replication of MVEV than KUNV

perhaps explain the more vigorous mononuclear cell infiltrates seen in the brains of

MVEV-infected susceptible HeJ and resistant mice DUB compared with KUNV-

infected mice (Section 6.2.1.1 and Section 6.2.2.1). In addition, the severity of brain

tissue inflammation was found to be more apparent in susceptible HeJ than resistant

DUB mice, again probably due to a much higher viral replication in the former mice.

From this study, it can be postulated that brain immunoinflammatory cells may play

different roles in different mouse strains. The greater degree of brain inflammation

suggests that immunopathological disease could contribute to the morbidity of

susceptible HeJ mice i.c. infected with either KUNV or MVEV. In contrast, the stronger

recruitment of brain mononuclear cells was associated with the recovery of resistant

DUB mice from i.c. MVEV infection.

The brain has evolved anatomically and physiologically to protect its delicate and vital

function from damaging pathogens or immune-mediated inflammation (Aloise, 2001).

However, the CNS is still constantly under an immune surveillance. Microglia form the

major APC in the CNS parenchyma and are capable of producing an array of cytokines

and chemokines in response to any insults or injury (Kreutzberg, 1996). Because of this,

microglia could also contribute to a development of immunopathological diseases since

the overproduction of cytokines and chemokines are toxic to the brain cells (Hanisch,

2001; Aloise, 2001). While contribution of microglia to CNS infection and disease

development has been documented in numerous studies, there is very little information

regarding the role of microglia in flavivirus infection. Recently, a robust increase of

activated microglia was demonstrated during i.c. infection with JEV in suckling

susceptible BALB/c mice (Ghoshal et al, 2007). Both in vivo and in vitro analysis has

implicated activated microglia in the neuronal death of these mice. Fatal encephalitis in

susceptible HeJ mice during i.c. infection with KUNV and MVEV also coincided with

high numbers of activated microglia/macrophages (Section 6.2.2.2). However, the role

of these cells in the pathogenesis of KUNV and MVEV in susceptible HeJ mice is yet to

be determined. Activated microglia/macrophages secrete a variety of substances that

can contribute to both neuroprotection and neurotoxicity in mice. These include

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

170

cytokines, glutamate, reactive oxygen species and NO (Hanisch, 2002; Streit, 2002).

Inhibition of NO activity has been shown to increase mortality in JEV infected mice

(Lin et al, 1997). Endogenously produced NO in monocytes and macrophages has also

been demonstrated to limit DENV replication (Neves-Souza et al, 2005). In contrast,

NO contributes to TBEV pathogenesis in mice as its inhibition improved the survival of

infected animals (Kreil and Eibl, 1995; 1996). In this laboratory, no difference in brain

NO production was observed between KUNV and MVEV infection in both susceptible

HeJ and resistant DUB mice compared with uninfected mice (Silvia et al, 2001; Shueb

2002). However, the lack of NO induction following i.c. KUNV and MVEV infection

did not eliminate the pathogenic role of microglia/macrophages during KUNV and

MVEV infection in HeJ mice since different effector molecules/substances such as IL-6,

IL-1β, TNFα and MCP-1 may be associated with KUNV and MVEV infection (Ghoshal

et al, 2007).

In resistant mice, brain microglia/macrophages were more activated on day 9 p.i. than

on day 5 p.i., as demonstrated by the increased staining intensity and more hairy

structure of these cells later in the infection. Interestingly, considerable numbers of

microglia/macrophages present in MVEV-infected DUB mice on day 9 p.i. compared

with KUNV-infected DUB mice, suggested that the increased accumulation of

microglia/macrophages may be involved in the clearance of MVEV infection. However,

additional studies are required to confirm this possibility, either by inhibition of

activation or selective depletion of microglia.

In conclusion, it was demonstrated here that the pathogenic mechanisms following

flavivirus infection, particularly with KUNV are different in susceptible versus resistant

mice even though both mouse strains develop fatal disease outcomes. The death in HeJ

mice after KUNV and MVEV i.c. challenge coincided with high brain viral titres,

severe brain inflammation and large numbers of activated microglia/macrophages, all of

which may have the ability to cause neuronal dysfunction without apparent brain cell

death. However, challenge of resistant DUB mice with the same viruses resulted in

significantly lower virus replication, less brain tissue inflammation and comparably

lower numbers of microglia accumulation than in susceptible HeJ mice. However, the

factors involved in such dramatically different outcomes of KUNV and MVEV

infections in resistant mice have not been identified. This study presented the evidence

that the abrogation of phenotypic expression of Flvr-like gene or an excessive immune

CHAPTER 6

KUNV AND MVEV INTRACEREBRAL INFECTION

171

response to virus infection did not contribute to the mortality in KUN-infected DUB

mice. However, the role of individual immune cell types in KUNV and MVEV

pathogenesis particularly in resistant mice is yet to be determined. The possibility that

KUNV may induce a greater accumulation and activation of specific immune cell types

remains to be explored. As will be described in the following chapter, further studies of

pathogenic mechanisms of KUNV and MVEV in susceptible HeJ and resistant DUB

mice targeting specific immune cell populations in the CNS was performed.

CHAPTER 7

IMMUNOPATHOLOGY

172

7.0 CHAPTER 7: ROLE OF CELL MEDIATED IMMUNITY IN

IMMUNOPATHOLOGY OR RECOVERY FOLLOWING

INTRACEREBRAL KUNV AND MVEV INFECTION IN MICE

7.1 INTRODUCTION

The immune system is a highly efficient defence mechanism that is capable of

combating any invading pathogen. The destructive potential of the immune system

however needs to be kept under strict control to prevent its harmful effect on the host

(Bachmann and Kopf, 2002). Deregulation of this control may result in a collateral

damage and could cause immunopathological diseases in the host. Many neurological

diseases are due to sustained, excessive or inappropriate immune response that exerts a

detrimental effect on the host.

The immune cells belonging to both branches of the host immune system, the adaptive

and innate immunity, have been reported to be involved in many immunopathological

diseases. For example, DHF has been suggested to occur due to antibody dependent

enhancement of virus infection of macrophages presumably by increasing the Fc

receptor-mediated internalisation of the virus (Halstead and O’Rourke, 1977).

Additionally, cross-reactive T cells have been demonstrated to contribute to DHF during

secondary infection, either by direct cytolysis or by production of cytokines

(Mongkolsapaya et al, 2006; reviewed in Fink et al, 2006). In the previous chapter,

death in susceptible HeJ mice was shown to be associated with high brain viral titres

following i.c. challenge with KUNV or MVEV. In addition, histological analysis of

infected-HeJ mouse brains suggested that a robust host immune response to flavivirus

infection in the CNS may exacerbate the course of infection, leading to fatal

encephalitis in these susceptible mice. However, at the start of this project, the

involvement of distinct host inflammatory cells in KUNV and MVEV pathogenesis in

susceptible HeJ mice was not known. On the contrary, since the development of fatal

disease in KUNV-infected resistant DUB mice was observed not at the peak of viral

replication, but rather at the time when infectious virus was almost cleared from the

brain, this precluded virus-induced direct cytopathology as the cause of death in

resistant DUB mice during i.c. challenge with KUNV. It was initially thought that the

death of resistant DUB mice following i.c. KUNV challenge was a result of an

CHAPTER 7

IMMUNOPATHOLOGY

173

excessive host immune response to the virus but as shown in the preceding chapter, no

severe tissue inflammation was observed in the brains of these mice. However, rather

than a massive tissue inflammation, the damaging effect of the immune response may

also caused by fine changes in the quantity or accumulation of particular immune cell

subpopulations in the CNS. Thus, the study described in this chapter was aimed at: (1)

performing a more detailed analysis on infiltrating brain leucocytes of infected mice in

order to identify and quantify different types of mononuclear cells accumulating in the

brains, particularly T cells, following KUNV and MVEV i.c. challenge and (2)

investigating whether T cells and cytokines secreted by these cells were associated with

the severity of disease during KUNV infection or recovery from MVEV infection in

resistant DUB mice. Since substantial evidence has linked T cells to either protection or

immunopathological diseases in virus-induced encephalitis (Binder and Griffin, 2003),

we hypothesised that the roles of T cells may vary following KUNV or MVEV infection

in different mouse strains. In susceptible HeJ mice, T cells may contribute to the

severity of diseases during both virus infections. However, in resistant DUB mice, T

cells are possibly linked to fatal outcomes of KUNV infection, yet may have a recovery

role following MVEV infection.

The analysis of different subpopulations of host immune cells included a flow

cytometry analysis to identify cells expressing surface markers for CD4+ and CD8+ T

cells as well as for B cells (CD19+) and microglia/macrophages (CD11b+) in brain

mononuclear cells isolated from infected mice. Studies on depletion of CD4+ or CD8+

T cells or both subsets of T cells prior to virus challenge, and the secretion of cytokines

locally in the brain and systematically in the circulation were also performed.

CHAPTER 7

IMMUNOPATHOLOGY

174

7.2 RESULTS

7.2.1 FLOW CYTOMETRIC ANALYSIS OF BRAIN MONONUCLEAR CELLS

FOLLOWING KUNV AND MVEV INFECTION

Histological analysis of HE stained brain tissue sections as described in Chapter 6

provided only a broad picture of brain inflammation and leucocytic infiltration.

However the role of individual cell types in the pathogenesis or recovery from KUNV

and MVEV infections, respectively, could not be ascertained from such study. In order

to obtain a better insight into the pathogenesis of these two viruses in terms of the types

and numbers of inflammatory cells attracted to the brains of infected mice, a more

targeted approach using flow cytometric analysis was used. To perform this experiment,

two groups of mice were infected i.c. with either KUNV or MVEV. At selected time

points of infection (see below), brains from three mice were harvested and pooled.

Following this, lymphocytes were isolated from the brains by discontinuous Percoll

gradient method and characterised using rat anti-mouse monoclonal antibodies against

cell surface markers for T lymphocytes subsets (CD4+ and CD8+), B (CD19+)

lymphocytes and macrophages/microglia (CD11b+). The total numbers of brain cells

isolated were determined using the Trypan blue exclusion dye assay prior to flow

cytometric analysis and these values were later used to calculate the total cell numbers

of different cell subtypes as shown in the following sections. Unless otherwise stated,

experiments described in this section were performed at least three times to allow for

the statistical analysis.

7.2.1.1 Analysis of cells infiltrating the brains of susceptible HeJ mice upon infection

with MVEV and KUNV

Using the approach described above, analysis of different immune cells present in the

brains of susceptible HeJ mice following virus infection was performed at the time of

death (day 5 p.i. and day 6 p.i. for KUNV and MVEV, respectively). Initial attempts

have been made to isolate brain mononuclear cells from susceptible HeJ mice 3 days

after KUNV or MVEV infection. However, very few cells were obtained which

coincided with the mild brain inflammation observed in these mice (Chapter 6).

Because of this, and as this study was more focused on resistant mice, experiment

planned to analyse brain infiltrates in these mice on day 3 p.i. was abandoned.

CHAPTER 7

IMMUNOPATHOLOGY

175

At the time of death, there were slightly more brain mononuclear cells isolated from

MVEV-infected than KUNV-infected HeJ mice (Table 7.1). This is in accordance with

the observations made in the preceding study (Chapter 6) concerning the stronger brain

inflammation observed in MVEV than in KUNV-infected mice.

As shown in Table 7.1, the predominant cell types present in the brains of infected mice

were microglia/macrophages (CD11b+) followed by T and B cells. Meanwhile, the

average number of CD4+ T cells found in brains of HeJ mice challenged with KUNV

was 9.4 ± 3.4 x 103 cells, whereas the number of the same T cell subset was double

following MVEV infection (Student t test, p < 0.04). Similarly, B cells and CD11b+

cells were also lower in the brains of KUNV-infected HeJ mice than that observed in

MVEV-infected HeJ mice. The difference in B cell numbers between these two

infections was significant (Student t test, p < 0.03). The higher numbers of CD11b+

cells found in the brains of MVEV-infected mice compared with KUNV-infected mice

did not correlate with the results from the previous study (Chapter 6) performed on

activated microglia. Perhaps this was because during immunohistochemical staining

using tomato lectin on brain tissue sections, only activated microglia/macrophages

present in the brains of infected mice were detected while resting

microglia/macrophages were excluded. In contrast, CD11b+ cell surface marker detects

both resting and activated microglia/macrophages.

Interestingly, KUNV induced a slightly higher numbers of brain infiltrating CD8+ T

cells (11.7 ± 2.5 x 103 cells) than MVEV (9.9 ± 1.4 x 10

3 cells) although these values

were not remarkably different (Student t test, p > 0.05). However, MVEV induced

higher numbers of total T cells than KUNV in the brains of susceptible HeJ mice. The

average total numbers of T cells recruited to the mouse brains at the time of death were

21.1 x 103 cells and 27.7 x 10

3 cells after KUNV and MVEV i.c. challenge,

respectively.

The findings above indicate that although similar lethal encephalitis was observed

following i.c. challenge of KUNV and MVEV in susceptible HeJ mice, interestingly,

these two viruses induced recruitment and extravasation of different numbers of

inflammatory cells to the brains of infected mice. Thus, death in KUNV and MVEV-

infected mice may be mediated by the different types of immune cells. It is possible that

CD8+ T cells contributed to the early death observed in HeJ mice during i.c. KUNV

CHAPTER 7

IMMUNOPATHOLOGY

176

challenge. However, further studies are required to confirm this possibility (see Section

7.2.2.2).

7.2.1.2 Analysis of lymphocytes in the brains and spleens of resistant DUB mice

following KUNV and MVEV infection

In this section, the infiltration and accumulation of brain mononuclear cells in resistant

DUB mice following i.c. infection with KUNV and MVEV was examined. These two

viruses have been shown previously (Chapter 4 and 6) to cause different outcomes of

infection in resistant DUB mice. Analysis of brain lymphocytes following virus

infection was carried out daily in resistant DUB mice from day 6 to day 9 p.i. In

addition, analysis of different numbers of mononuclear cells was also performed on the

spleens of infected mice from day 5 to day 9 p.i. As shown in Figure 7.1, the total

numbers of splenocytes decreased over time following both virus infections in resistant

DUB mice, although there were more cells present in the spleens of mice infected with

MVEV than KUNV. The total numbers of splenocytes were significantly different

following challenges with these two virus on days 7 (Student t test, p < 0.02,) and 8 p.i.

(Student t test, p < 0.05). Additionally, on day 8 and 9 p.i, spleens harvested from

KUNV-infected resistant mice had decreased in size and were very small in comparison

to those isolated from MVEV-infected mice. It is not known why this occurred,

although it could be speculated that spleen cells in KUNV-infected mice underwent

apoptosis or were recruited to the brain.

In contrast, brain leucocytes derived from resistant mice infected with KUNV or MVEV

increased in numbers when monitored from day 6 to day 9 p.i. (Figure 7.1). In

agreement with the histopathological study performed in the previous section (6.3.1),

there were consistently more leucocytes isolated from the brains of MVEV-infected

than KUNV-infected resistant mice throughout the course of disease, although these

values were not significantly different (Student t test, p > 0.05).

CHAPTER 7

IMMUNOPATHOLOGY

177

Table 7.1. Number of brain infiltrating leucocytes isolated from HeJ mice that

succumbed to KUNV and MVEV infection.

Total

isolated

brain cells

(x 105)

CD4 T cells

(x 103)

CD8 T cells

(x 103)

B cells

(x 103)

CD11b cells

(x 103)

KUNV 12.2 ± 0.2 9.4 ± 3.4 11.7 ± 2.5 5.9 ± 0.2 542.0 ± 111.8

MVEV 12.4 ± 0.2 17.7 ± 3.3 9.9 ± 1.4 9.0 ± 1.2 781.0 ± 50.0

Susceptible HeJ mice were i.c. infected with 1.7 x 105 i.u. of KUNV or 3.4 x 10

3 i.u. of

MVEV. Brains were harvested from HeJ dying from KUNV or MVEV infection on day

5 and 6 p.i., respectively. Average number of brain cells was derived from 3 mice. Data

shown were average number of cells ± standard errors.

CHAPTER 7

IMMUNOPATHOLOGY

178

In addition to determining the total number of mononuclear cells as reported above, the

numbers of individual cell types, T cells (CD4+ and CD8+), B cells and

microglia/macrophages (CD11b+) were also determined in both the spleens and brains

of infected resistant DUB mice. In general, infection with both viruses resulted in

diminished numbers of cells for all cell types being analysed in the spleens as the

disease progressed. B cells were the most predominant cell type found in the spleens,

followed by CD4+ T cells. However, both T cells subsets, B cells and CD11b+ cells

were demonstrated to be more numerous in the spleens of MVEV-infected mice that

KUNV-infected mice (Figure 7.2). This coincided with the greater numbers of total

cells obtained from these mice compared to those infected with KUNV.

In contrast to that observed in the spleens, CD11b+ cells were the principal

inflammatory cells recruited/residing in the mouse brains following both virus

infections. This is because in addition to recruited macrophages, microglia which are

unique to the brain, express CD11b+ cell surface marker as well. This was followed by

the cells carrying CD8+ cell surface markers. CD11b+ cells were 14-17 times more

numerous than CD8+ cells on day 6 p.i., and 4-9 times on days 7 p.i. to 9 p.i. with

KUNV and MVEV infection. The numbers of brain CD11b+ cells increased to a great

extent from day 6 p.i. to day 7 p.i. in mice infected with either virus. From day 7 p.i. to

day 9 p.i., the numbers of these cells did not change considerably. However, MVEV

induced a greater recruitment of CD11b+ cells to mouse brains than KUNV, although

the difference was not significant at any time point p.i. In addition to macrophages and

microglia, dendritic cells also express CD11b+ cell surface marker. Since presence of

dendritic cells in the brain parenchyma during CNS diseases has been documented

(reviewed in McMahon et al, 2006; Fisher et al, 2001; Fisher et al, 2000), a small scale

study was conducted to investigate whether or not dendritic cells were the major cell

type present in CD11b+ cell population.

CHAPTER 7

IMMUNOPATHOLOGY

179

A Spleen

0

10

20

30

40

50

60

70

80

90

5 6 7 8 9

Days post infection

Nu

mb

er

of

cells (

x10

5)

B Brain

0

2

4

6

8

10

12

14

6 7 8 9

Days post infection

Nu

mb

er

of

cells (

x10

5)

KUN MVE

Figure 7.1. Total number of cells isolated from (A) spleens and (B) brains of

resistant mice challenged i.c. either with KUNV or MVEV.

At different time points (5 to 9 days) after infection, spleens and brains were harvested

from 3 mice and homogenised to obtain cell suspension. Trypan blue exclusion dye was

used to enumerate total number of cells. Spleens were harvested from 3-5 mice and cells

were analysed individually. In contrast, brain cells were pooled from 3 mice and

analysed as one sample. Experiment on brain cells were repeated 3 times. Data

presented as average number of cells ± SE.

CHAPTER 7

IMMUNOPATHOLOGY

180

A CD4

0

5

10

15

20

25

5 6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

6)

B CD8

0

5

10

15

20

25

5 6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

6)

0

2

4

6

8

10

12

14

6 7 8 9

Days post infection

Num

ber

of

ce

lls (

x1

05)

KUNV MVEV

Figure 7.2. Flow cytometric analysis of splenocytes in DUB mice following i.c.

KUNV and MVEV infection.

A) CD4+ T cells B) CD8+ T cells C) B cells D) CD11b+ cells. Data shown are average

values from 3 separate experiments ± standard error. At days 5 to 9 p.i., spleens were

removed and total cells were isolated and enumerated. Aliquots containing 106 cells

were labelled with antibodies specific for CD4+, CD8+, CD19+ (B cells) and CD11b+

cell surface markers. Flow cytometric analysis was later performed and 104 events were

recorded.

CHAPTER 7

IMMUNOPATHOLOGY

181

C B cells

0

5

10

15

20

25

30

35

40

45

50

5 6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

6)

D CD11b+

0

2

4

6

8

10

12

14

5 6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

6)

0

2

4

6

8

10

12

14

6 7 8 9

Days post infection

Num

ber

of

cells

(x10

5)

KUNV MVEV

CHAPTER 7

IMMUNOPATHOLOGY

182

This study was only performed on brain mononuclear cells harvested from resistant

DUB mice 9 days after i.c.challenge with either KUNV or MVEV on day 9 p.i.

Dendritic cells express both CD11b+ and CD11c+ cell surface markers and therefore

dual staining was performed on mononuclear cells using these two cell surface markers

in order to identify dendritic cells. It was found that dendritic cells only comprised

between 6-10% of total CD11b+ cells (data not shown) and thus were not the major cell

type of the brain’s CD11b+ cells in infected-esistant DUB mice.

The second largest population of infiltrating leucocytes found in the brains of resistant

DUB mice during KUNV and MVEV i.c. challenge was CD8+ T cells. DUB mice

infected with MVEV showed a sharp increase of CD8+ T cell numbers from 9.8 x 103

cells on day 6 p.i. to 127 x 103 cells on day 7 p.i. The numbers of CD8+ T cells

recruited to the brains of MVEV-infected DUB mice peaked on day 7 p.i. Following

this, CD8+ T cells gradually decreased, coinciding with the clearance of infectious

virus, and reached a low level on day 9 p.i (64.6 x 103 cells) (Figure 7.3B). Similarly,

KUNV infection resulted in the CD8+ T cell recruitment to the brain that reached a

peak on day 7 p.i. (156 x 103 cells). Interestingly, KUNV consistently induced higher

numbers of brain CD8+ T cells than MVEV in resistant mice from days 7 to 9 p.i.

Additionally, CD8+ T cells persisted at relatively high numbers on day 9 p.i. (143 x 103

cells) when mice started to succumb to fatal infection. The numbers of brain CD8+ T

cells on day 9 p.i. following KUNV infection were significantly different than that

observed after non-fatal MVEV infection in the same mouse strain (Student t test, p <

0.03). The prolonged presence of CD8+ T cells in the brains of sick KUNV-infected

DUB mice coincided with the incomplete clearance of infectious virus found in these

mice as detected by TCID50 bioassay. Thus, this study showed that from day 7 p.i. to

day 9 p.i., the numbers of CD8+ T cells positively correlated with the brain viral titres.

Furthermore, although MVEV induced a stronger overall brain inflammation, KUNV

caused a greater CD8+ T cells response and this subtype of T cells may have important

contribution in KUNV pathogenesis and subsequent severe infection in DUB mice.

CHAPTER 7

IMMUNOPATHOLOGY

183

A CD4

0

50

100

150

200

250

6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

3)

B CD8

0

50

100

150

200

250

6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

3)

0

2

4

6

8

10

12

14

6 7 8 9

Days post infection

Num

ber

of

ce

lls (

x1

05)

KUNV MVEV

Figure 7.3. Analysis of brain infiltrating leucocytes in DUB mice following KUNV

and MVEV infection.

A) CD4+ T cells B) CD8+ T cells C) B cells D) CD11b+ cells. At days 6 to 9 p.i.,

brains mononuclear cells were isolated and enumerated. Aliquots containing 106 cells

were labelled with antibodies specific for CD4+, CD8+, CD19+ (B cells) and CD11b+

cell surface markers. Flow cytometric analysis was later performed and 104 events were

recorded. *The number of cells was significantly different between KUNV and MVEV

infection. Data shown are average values from 3 separate experiment ± SEM. In each

separate experiment, 3 brains were harvested and pooled.

*

*

CHAPTER 7

IMMUNOPATHOLOGY

184

C B cells

0

10

20

30

40

50

60

70

6 7 8 9

Days post infection

Nu

mb

er

of

ce

lls

(x

10

3)

D CD11b+

0

100

200

300

400

500

600

700

800

6 7 8 9

Days post infection

Nu

mb

er

of

cell

s (

x10

3)

0

2

4

6

8

10

12

14

6 7 8 9

Days post infection

Num

ber

of

cells

(x10

5)

KUNV MVEV

*

CHAPTER 7

IMMUNOPATHOLOGY

185

Other cell types that were analysed showed the opposite pattern of

recruitment/accumulation to that demonstrated by CD8+ T cells. The numbers of

CD4+T cells in general, increased as the infection with both viruses progressed (Figure

7.3A). However, higher numbers of CD4+ T cells were observed between day 7 to day

9 days after MVEV infection than following KUNV infection. On day 8 p.i., the

difference between the recruited CD4+ T cells in KUNV and MVEV-infected resistant

DUB mice was significant (69 x 103 cell and 104 x 10

3 after KUNV and MVEV

infection, respectively; Student t test, p < 0.005).

The numbers of B cells increased throughout the infection and reached the highest

levels on day 9 p.i. during MVEV infection in DUB mice. In contrast, although

increased numbers of B cells were also observed in the brains of KUNV-infected DUB

mice initially, the numbers of these cells declined on day 9 p.i. when mice succumbed to

the infection. Similar to CD4+ T cells, MVEV infection also caused higher levels of B

cells recruitment into the brains of infected DUB mice compared to KUNV infection,

with the greatest difference demonstrated on day 9 p.i. (Student t test, p < 0.05).

The ratio of brain CD8+ to CD4+ T cells in MVEV-infected DUB mice on days 6, 7, 8

and 9 p.i. were 1:1.1, 2:1, 1:1 and 1:1.4 respectively. Thus, except on day 7 p.i. (when

CD8+ T cells were doubled in comparison to CD4+ T cells), the numbers of CD8+ T

cell and CD4+ T cells recruited to MVEV-infected animals were almost equivalent. In

contrast, CD8+:CD4+ T cells in resistant mice challenged with KUNV on days 6, 7, 8

and 9 p.i. were 1:1.1, 2.9:1, 1.8:1 and 1.7:1 respectively. This clearly demonstrated that

KUNV infection resulted in greater numbers of CD8+ T cells being recruited into the

brains (almost 2-3 times more) than CD4+ T cells from day 7 p.i. onwards. When the

total numbers of T cells were calculated, the numbers of brain T cells in DUB mice

infected with either virus were similar on day 6 p.i. However, on day 7 p.i., there were

on average 210.5 x 103 T cells following KUNV infection and 189.34 x 10

3 T cells

following MVEV infection. In the next 2 days, the total numbers of T cells following

KUNV infection remained relatively stable (197.7 x 103 cells and 225.6 x 10

3 cells on

day 8 and 9 p.i. respectively). In contrast, following MVEV challenge, T cell numbers

increased to 213.25 x 103 cells on day 8 p.i. before declining to 157.52 x 10

3 cells on

day 9 p.i. Significantly different numbers of T cells was observed in the brains of DUB

mice infected with KUNV compared with MVEV on day 9 p.i. (213.2 x 103 and 157.5

x 103 cells during KUNV and MVEV infection, respectively, Student t test, p < 0.05).

CHAPTER 7

IMMUNOPATHOLOGY

186

As presented in this section, the relative proportion of different inflammatory cell sub-

populations in response to i.c. infection with KUNV and MVEV as determined by flow

cytometry provided a better insight in the quality of inflammatory response elicited by

these viruses. This study has demonstrated that the different proportions of

immunoinflammatory cells recruited to the brain possibly contributed to the different

outcomes of infection observed during KUNV and MVEV i.c. challenge in resistant

DUB mice. While more microglia/macrophages, CD4+ T cells and B cells were present

in the brains of MVEV-infected DUB mice, greater numbers and sustained response of

CD8+ T cells were observed following KUNV challenge. This finding is very crucial as

it showed for the first time that CD8+ T cells may have immunopathological role

following KUNV infection.

7.2.1.3 Analysis of MHC cell surface up-regulation on brain CD11b+ cells following

flavivirus infection.

MHC class I and II molecules play important role in the adaptive immune responses as

these molecules participate in the activation of CD8+ and CD4+ T cells, respectively

(Abraham and Manjunath, 2006). MHC I class I molecules are required for CD8+ CTL

cells to exert their effector function (Dorries, 2001). Unlike other viruses that evade host

immune surveillance by down-regulating MHC class I expression on infected cells,

flaviviruses are known to up-regulate expression of these molecules (King et al, 2003).

In order to further elucidate the pathogenesis of KUNV and its effect on the sustained

CD8+ T cell response, the ability of KUNV to up-regulate cell surface expression of

MHC antigen particularly MHC class I was examined. The main objective of this study

was to compare the levels of MHC molecules up-regulation by KUNV versus MVEV as

possible mechanism for the sustained T cell response observed in KUNV infection.

However, the MHC cell surface molecules analysis was only conducted on CD11b+

cells derived from resistant mouse brains infected with KUNV or MVEV. Only these

cells were chosen as they are known to possess the antigen presenting properties in the

brain and interact with T cells (Hanisch, 2002). Additionally, they could potentially be

infected with flavivirus.

The MHC class I molecules expression can be induced or up-regulated in most cells

following virus infection or exposure to cytokines (King et al, 2004). Thus, MHC class

CHAPTER 7

IMMUNOPATHOLOGY

187

I molecules analysis was initially planned to be performed in parallel with KUNV or

MVEV immunostaining using virus specific mouse anti-NS1 antibodies. This

experiment would help to provide better information on whether CD11b+ cells support

flavivirus infection in vivo, in addition to neurons which are the major permissive cells

for flaviviruses in the brain. Furthermore, the dual staining would also help to determine

whether or not the up-regulation of MHC I molecules on CD11b+ cells is induced

directly by virus infection. Unfortunately, initial flow cytometry analysis of CD11b+

stained cells with anti-NS1 antibodies failed because of a high background problem.

Due to a time constraint and limited reagents available, this dual immunostaining could

not be optimised and had to be abandoned. Alternatively, the flavivirus infection was

monitored in the total mononuclear cells containing CD11b+ cells as well as other

inflammatory cells that were isolated from HeJ mice and cultured ex-vivo for a day to

monitor the virus release in the cell culture supernatant. This method successfully

showed the presence of infectious virus at low titres in total brain mononuclear cells

when assayed by TCID50 bioassay (data not shown). However, this approach did not

target specifically CD11b+ cells, so further CD11b+ cell sorting prior to ex-vivo

cultivation could be introduced for a more specific analysis of virus infection in

microglia/macrophage cell population.

To study the up-regulation of MHC molecules on CD11b+ cells, brains from three

resistant mice infected either with KUNV or MVEV were harvested on different days.

Mononuclear cells were isolated from pooled brains (3 brains) prior to simultaneous

staining with the rat anti-mouse CD11b+ and mouse anti-mouse MHC class Ia

monoclonal antibodies. Flow cytometry was used to analyse the MHC class I

upregulation. As shown in Figure 7.4, CD11b+ cells from brains of uninfected mice did

not show detectable levels of MHC I and II molecules. The up-regulation of MHC I

molecules was observed 5 days after infection. Interestingly, the expression of these

molecules was greater in the cells derived from KUNV (mean fluorescence intensity,

MFI 29.4; data not shown) than in the cells isolated from MVEV-infected resistant

DUB mice (MFI 8.8, data not shown). A greater level of MHC I expression in

CD11b+ cells following KUNV infection than in infection with MVEV was seen

throughout the course of infection. The MFI increased 2 days later in both infections

(54.4 and 48.5 following KUNV and MVEV infection, respectively,) and then slightly

decreased on day 9 p.i. (49.6 and 38.8 after KUNV and MVEV infection, respectively).

CHAPTER 7

IMMUNOPATHOLOGY

188

In addition to MHC class I, the expression level of MHC II cell surface molecules was

also examined. MHC class II expression was found to be also up-regulated in CD11b+

cells isolated from resistant DUB mice following either KUNV or MVEV infection.

The kinetics of expression of MHC II molecules was similar to that observed in the

MHC I up-regulation. Minimal expression was observed on day 5 p.i. and it then

increased by day 7 p.i. However, on day 9 p.i., the expression of MHC II molecules on

CD11b+ cells was slightly reduced after both KUNV and MVEV infections. Similar to

what observed with MHC class I expression, KUNV also induced slightly higher levels

of MHC class II expression than MVEV (data not shown).

The data presented above indicate that KUNV elicits a slightly stronger up-regulation of

the MHC molecules than MVEV on CD11b+ cells when monitored from days 5 to 9 p.i.

Although this experiment could not directly determine the infectibility of CD11b+ cells,

it shows correlation between the upregulation of MHC molecules expression and the

course of in vivo infection, particularly with KUNV. The continuous up-regulation of

MHC class I on CD11b+ cells indicates that the interaction of T cells and CD11b+ cells

in antigen-dependent manner is possible, thus explaining the sustained CD8+ T cell

response observed in brains of KUNV-infected DUB mice.

7.2.2 T CELL DEPLETION STUDIES

Flow cytometry analysis demonstrated that different types of inflammatory cells were

recruited to the brains of infected mice. While higher numbers of brain CD8+ T cells

were found in KUNV-infected HeJ and DUB mice, MVEV-infected mice showed a

greater presence of CD4+ T cells, B cells and microglia/macrophages. This data

implicates CD8+ T cells involvement in the severe outcomes of KUNV infection

particularly in resistant DUB mice.

Several recent studies have been carried out to determine the involvement of T cells in

flavivirus pathogenesis. These studies showed that T cells may have different roles,

depending on virus types, route of infection and mouse strain used (Wang et al, 2003b;

Shrestha and Diamond, 2004). T cells may either be beneficial to the host or they may

CHAPTER 7

IMMUNOPATHOLOGY

189

MHCI MHCII

100

101

102

103

104

FL1-H: FITC

0

20

40

60

80

100

10

010

110

210

310

4

FL1-H: FITC

0

20

40

60

80

100

100

101

102

103

104

FL1-H: FITC

0

20

40

60

80

100

10

010

110

210

310

4

FL1-H: FITC

0

20

40

60

80

100

100

101

102

103

104

FL1-H: FITC

0

20

40

60

80

100

10

010

110

210

310

4

FL1-H: FITC

0

20

40

60

80

100

100

101

102

103

104

FL1-H: FITC

0

20

40

60

80

100

10

010

110

210

310

4

FL1-H: FITC

0

20

40

60

80

100

Figure 7.4. Up-regulation of MHC class I and II molecules in CD11b+ cells

following KUNV and MVEV infection.

Da

y 5

p.i

. D

ay

7 p

.i.

Da

y 9

p.i

. U

nin

fecte

d

Negative control

Uninfected brain

KUNV

MVEV

CHAPTER 7

IMMUNOPATHOLOGY

190

have detrimental effects and contribute to the development of fatal encephalitis. To

further clarify the role of T cells in KUNV and MVEV infection as well as whether this

role would be similar in susceptible HeJ mice and resistant DUB mice, T cells depletion

was performed using anti-CD4+ or CD8+ monoclonal antibodies.

7.2.2.1 Pilot study to determine the optimum antibody depletion time

A small scale pilot study was initially conducted to determine the optimum time for T

cells depletion (data not shown). Two groups of five DUB mice received either 100uL

of anti CD8+ T cell antibodies on days -2, 0, 2, 6 and 8 after KUNV infection, or on

days 5, 7 and 9 p.i. following KUNV infection. A positive control consisting of five

mice infected with KUNV only was also included and development of disease in all

mice was monitored. Following infection, all mice in the control group began to show

signs of sickness such as hunched posture and ruffled fur on day 7 p.i. that became more

apparent on day 8 p.i. By day 9 p.i., these animals were culled as they were very sick

and some of them developed hind leg paralysis. In contrast, KUNV-infected DUB mice

that received anti CD8+ antibodies on -2, 0, 2, 6 and 8 days p.i. only started to have

mild ruffled fur on day 10 p.i. By day 12 p.i., 4 of the mice died. The last remaining

mouse developed fatal disease by day 14 p.i. The signs of disease and development of

fatal encephalitis was also shown to be delayed in the group that received depleting

antibody on days 5, 7 and 9 p.i. These mice only became slightly sick on day 8 p.i. and

on day 10 p.i., three mice succumbed to virus infection. The rest of the mice had fatal

disease outcomes on day 14 p.i. The ATD for different groups of mice were as

followed: day 9 ± 0 p.i. for mice that were infected with virus only, day 12.7 ± 0.7 p.i.

for mice that received antibodies before infection and 11.7 ± 1.2 p.i. for animals that

were treated with depleting antibodies after infection. Since mice treated with depleting

antibodies prior to infection survived much longer than other groups of mice, in the

subsequent studies, anti-CD4+ and CD8+ T cells antibodies were given to resistant

DUB mice on days -2, 0, 2, 6 and 8 p.i. In susceptible HeJ mice, since they succumbed

to KUNV and MVEV infection 5 and 6 days after infection, respectively, anti-CD4+

and CD8+ antibodies were administered on days -2, 0, 2 and 4 p.i.

To confirm depletion of T cells, separate groups of resistant DUB mice were treated

with depleting antibodies on day 1 and day 3 (150uL CD4+ antibodies or 100uL CD8+

antibodies). The concentration was 600 µg/ml and 926 µg/ml for anti-CD4+ and anti

CHAPTER 7

IMMUNOPATHOLOGY

191

Figure 7.5. Analysis of T cells depletion in DUB mice by flow cytometry after

treatment with cytotoxic anti-CD4 and anti-CD8 antibodies.

A) CD8+ T cells composition in normal mouse spleen. B) CD8+ T cells were depleted

following antibody treatment. C) CD4+ T cells composition in normal mouse spleen D)

CD4+ T cells were depleted following antibody treatment. Mice were given 150 L

cytotoxic anti CD4 or 100 µL CD8 culture supernatant on day 1 and day 3 and spleens

were harvested on day 4 and analysed whether depletion had occurred. 96% and 95%

depletion of CD4+ and CD8+ T cells was achieved, respectively following treatment

with corresponding antibodies.

10 0 10 1 10 2 10 3 10 4

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide

Sca

tte

r

7.03

10 0 10 1 10 2 10 3 10 4

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide

Sca

tte

r

0.34

10 0 10 1 10 2 10 3 10 4

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide

Sca

tte

r

0.79 22

10 0 10 1 10 2 10 3 10 4

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide

Sca

tte

r

A

100

101

102

103

104

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide S

catt

er

6.83

C

100

101

102

103

104

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide S

catt

er

6.83

D

100

101

102

103

104

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide S

catt

er

6.83

CD8 CD8

CD4 CD4

B

100

101

102

103

104

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide S

catt

er

6.83

B

100

101

102

103

104

FL4-H: Cy5

0

200

400

600

800

1000

SS

C-H

: S

ide S

catt

er

6.83

CHAPTER 7

IMMUNOPATHOLOGY

192

CD8+ culture supernatant, respectively. Then on day 4, splenocytes were harvested and

labeled with rat anti-mouse anti-CD8+ (3.11M) and anti-CD4+ (RL174) monoclonal

antibodies that recognised different epitopes on T cells than those used to deplete CD4+

and CD8+ T cells . By flow cytometric analysis, 95% and 96% depletion was achieved

following treatment with anti CD8+ and CD4+ antibodies, respectively, (Figure 7.5).

Depletion of T cell subsets is a transient process as shown by the repopulation of the

spleens as well as the brains with T cells at later time points post infection. Flow

cytometric analysis of MVEV-infected mice, showed that T cells started to infiltrate the

brain as early as 14 days after infection, which was eight days after the antibody

treatment with anti-CD4+ and CD8+ antibodies ceased (data not shown).

7.2.2.2 Effect of CD4+ or CD8+ T cells depletion on mortality following flavivirus

infection in susceptible mice

This part of the study was aimed at investigating the contribution of T cells to the

pathogenesis of KUNV and MVEV in susceptible HeJ mice. Two groups of five

susceptible HeJ mice received a treatment with anti-CD4 antibodies. Another two

groups consisting of 15 HeJ (pooled from two separate experiments) received a

treatment with anti-CD8 antibodies prior to i.c. challenge with either KUNV or MVEV.

In addition, 10 HeJ mice infected with viruses only were also included as a control

group.

Depletion of CD4+ T cells caused a slight increase in the ATD values with no effect on

mortality rate following both virus infections. While all control HeJ mice died on day 5

or day 6 p.i. after KUNV or MVEV challenge, respectively, only 80% of mice

following CD4+ T cell depletion died on the same day as the control mice while the

other 20% died on the next day (Figure 7.6). The ATD and brain viral titres however did

not differ significantly in these mice (Table 7.2). This study indicated that CD4+ T cells

may only have a slight but not important contribution to the development of fatal

encephalitis in HeJ mice.

Depletion of CD8+ T cells showed different effects on the course of infection of KUNV

versus MVEV in susceptible HeJ mice. All susceptible HeJ mice lacking CD8+ T cells

succumbed to KUNV challenge on the same day as the control mice. Brain viral titres as

CHAPTER 7

IMMUNOPATHOLOGY

193

shown by TCID50 bioassay were also not significantly different between these two

groups of mice. Interestingly, depletion of the same T cell subset seemed to exacerbate

the course of MVEV infection in susceptible mice. Susceptible HeJ mice did not only

showed signs of diseases, including ruffled hair, hunched back and flaccid tail earlier

than the control group (day 4 p.i.), but 10 out of 15 mice also died on day 5 p.i. while

the control group succumbed to the infection on day 6 p.i. The shorter ATD in MVEV-

infected HeJ in the absence of CD8+ T cells (statistically significant, Student t test, p

<0.01) demonstrated that this subset of T cells possibly has a neuroprotective role

following MVEV infection in susceptible HeJ mice. This is in contrast to the lack of

CD8+ T cell depletion effect on the mortality rate of i.c. KUNV-infected susceptible

HeJ mice (Figure 7.6 and Table 7.2). However, mortality rate did not change in MVEV-

infected HeJ mice that lacked CD8+ T cells which indicated that CD8+ T cells have

only a minor contribution to the development of fatal encephalitis in these mice.

From the data obtained, it can be concluded that T cells do not significantly contribute

to the pathogenesis of KUNV or MVEV in susceptible HeJ mice. Furthermore, CD8+ T

cells may exert some protection to susceptible HeJ mice only during i.c. MVEV

infection which is interesting since KUNV induced more CD8+ T cells than MVEV in

the brains of susceptible HeJ mice.

7.2.2.3 Effect of CD4+ or CD8+ T cells depletion on mortality following flavivirus

infection in resistant DUB mice

The role of T cell subsets in flavivirus pathogenesis in susceptible HeJ and resistant

DUB mice has never been studied comparatively and therefore it is not known whether

T cells would assume similar roles in susceptible versus resistant mice following the

same virus infection. However, the effect of similar depletion on survival/mortality rate

was expected to be greater in resistant DUB mice following virus infection. The reasons

for this were 1) the much slower course of virus infection in DUB mice 2) the greater

recruitment of T cells into the brains of these mice and finally 3) the sustained

accumulation of T cells in KUNV-infected resistant mice as a possible cause of the poor

outcome of infection.

CHAPTER 7

IMMUNOPATHOLOGY

194

0

20

40

60

80

100

1 2 3 4 5 6 7

Days post infection

Su

rviv

al

(%)

KUNV

KUNV-CD4

KUNV-CD8

MVEV

MVEV-CD4

MVEV-CD8

Figure 7.6. Effect of CD4 or CD8 cells depletion on mortality following KUNV and

MVEV infection in flavivirus susceptible HeJ mice.

Fifteen mice were used for each group except for groups that received CD4 antibodies,

where only 5 mice were used per group.

Table 7.2. Effect of CD4+ and CD8+ T cells depletion on mortality of HeJ mice

following i.c. challenge with KUNV or MVEV.

Control CD4

depletion

CD8

depletion

KUNV

Survival (%) 0

(0/10)a

0

(0/5)

0

(0/15)

ATDb ( day p.i) 5.0 0.0 5.0 0.3 5.0 0.0

Average viral titres c

(Log10 TCID50/ 0.01g

tissue) 7.8 0.5 7.6 0.5 7.2 0.5

MVEV

Survival (%) 0

(0/10)

0

(0/5)

0

(0/15)

ATDb ( day p.i) 6.0 0.0 6.0 0.5 5.0 0.5

Average viral titres c

(Log10 TCID50/ 0.01g

tissue) 9.8 0.6 9.2 0.3 9.8 0.4

a Number of animals died per total number of animal

b Average time to death

cVirus titres were taken from mice that succumbed to the infection

CHAPTER 7

IMMUNOPATHOLOGY

195

As shown by Figure 7.7, i.c. challenge in 8-10 weeks old resistant DUB mice with

KUNV resulted in all 15 mice developing fatal encephalitis. A very mild signs of

disease was seen on day 6 p.i. in some mice which later became more evident on day 7

p.i. Death was first recorded in three mice on day 8 p.i., followed by another nine mice

on day 9 p.i, and the remaining of mice died on day 10 p.i. The ATD was day 9.0 ± 1.0

p.i. while average brain titres was 0.6 ± 1.0 log10 TCID50/0.01g for these mice (Table

7.3). In contrast, resistant DUB mice depleted of CD4+ T cells prior to KUNV infection

did not show signs of sickness until day 7 p.i., which was a day later than observed in

control KUNV-infected mice. The mice also started to succumb later than the control

infected mice. Two mice succumbed to KUNV infection 9 days after infection. More

deaths were observed on the following days; seven deaths occurred on day 10 p.i. and

four more deaths were reported on day 11 p.i. By day 12 p.i., all mice developed fatal

disease outcomes and had to be euthanised. The ATD was 10.4 ± 0.2 p.i. days, which

was significantly greater than in control infected DUB mice (ATD was 9.0 ± 1.0 p.i.

days Student t test, p < 0.01). The average brain virus titres in CD4+ T cell-depleted

mice succumbing to KUNV infection was 4.6 ± 0.7 log10 TCID50/0.01g, which was four

logs higher than found in control mice (Student t test, p < 0.006) (Table 7.3). This

finding is interesting since the numbers of recruited CD4+ T cells were lower in the

brains of KUNV-infected DUB mice compared to MVEV-infected DUB mice (Section

7.2.1.2) and it was initially thought that CD4+ T cells may not have detrimental effect

in KUNV-infected DUB mice.

Depletion of CD8+ T cells had a much greater effect on the development of disease and

survival of DUB mice following i.c. KUNV infection than depletion of CD4+ T cells. In

general, development of disease was very slow and gradual compared to control

infected mice and CD4+ T cells-depleted mice. Some of resistant DUB mice lacking

CD8+ T cells exhibited severe ruffled fur and hunched back only 9 days following

infection. One death was recorded the following day, which was 2 days later than the

control mice. Later, a few of the remaining mice also developed fatal encephalitis and

by day 18 p.i., only 9 out of 15 mice (60%) succumbed to the infection while the rest

(40%) survived (Kaplan Meier test, p < 0.007 when compared to control infected DUB

mice). CD8+ T cell-depleted mice that died from KUNV infection had an ATD of 12.4

± 0.7 p.i. days which was remarkably longer than in the control group (Student t test, p

< 0.001). Brain viral titres determined in the same mice were also much higher than

the control group (3.3 ± 1.4 log10 TCID50/0.01g, Student t test, p < 0.03).

CHAPTER 7

IMMUNOPATHOLOGY

196

Surprisingly, depletion of CD4+ or CD8+ T cells did not affect the survival of resistant

DUB mice i.c. infected with MVEV (Figure 7.7). As T cells are involved in virus

clearance during flavivirus infection (Shrestha and Diamond, 2004), it was expected

that T cell depletion would affect the resistance of DUB mice to MVEV. However,

resistant DUB mice did not exhibit any signs of disease and survived the infection,

similar to control resistant DUB mice during a 30-days monitoring period. This finding

clearly suggests that CD4+ or CD8+ T cells alone do not play a major role in the

survival of resistant DUB mice during MVEV infection and that the Flv gene may have

a greater contribution to the outcome of infection.

Although the depletion of either subset of T cells did not cause morbidity or abrogation

of resistance to MVEV in DUB mice, we hypothesised that virus titres in the brains of

infected DUB mice may have been affected in the absence of T cells. In order to test

this hypothesis, virus titres in the brains of MVEV-infected DUB mice lacking either

CD4+ or CD8+ T cells were determined in a separate experiment. The brains were

harvested from 3 mice in each group on days 5, 7, 9, 11 and 14 days p.i. and viral titres

assayed by TCID50 bioassay. As shown in Figure 7.8, on day 5 p.i. MVEV-infected

resistant mice showed similar brain viral replication in the presence or absence of

functional CD4+ and CD8+ T cells. However, after day 5 p.i., the brain viral titres in

control DUB mice i.c. infected with MVEV decreased sharply, and by day 9 p.i.,

infectious virus was no longer detected. In contrast, virus production in MVEV-infected

resistant DUB mice that lacked either CD4+ or CD8+ T cells did not decline and

remained relatively the same on day 9 p.i. (Figure 7.8, Table 7.3). The inability of these

mice to clear MVEV infection from the brains late in infection (day 9 p.i.) demonstrates

the pivotal participation of CD4+ and CD8+ T cells in clearing MVEV. However, after

day 9 p.i., MVEV replication in the brains of resistant DUB mice with transiently

impaired CD4+ or CD8+ T cells started to decline and on day 14 p.i., infectious MVEV

was no longer detectable by TCID50. This coincided with the repopulation of CD4+ and

CD8+ T cells observed in the spleens by day 14 p.i. (Section 7.2.2.1) as (data not

shown) as this T cell depletion is a transient process.

The depletion of T cell subsets studies provided the evidence for the first time on the

role of CD4+ and CD8+ T cells in the pathogenesis of KUNV in resistant DUB mice. In

contrast, CD4+ or CD8+ T cells alone were not neuroprotective as the absence of either

CHAPTER 7

IMMUNOPATHOLOGY

197

cell did not have an impact of the well being of MVEV-infected DUB mice despite the

slower clearance of infectious virus in the brains of these mice.

7.2.2.4 Effect of total T cells (CD4+ and CD8+) depletion on mortality following

KUNV and MVEV infection in resistant DUB mice

Since a single depletion of either CD4+ or CD8+ T cells did not affect resistant DUB

mice following i.c. MVEV challenge, the depletion effect of both CD4+ and CD8+ T

cells on mouse survival was examined. Two groups of 15 resistant DUB mice were

given both anti-CD4+ and CD8+ antibodies on days -2, 0, 2, 6 and 8 p.i. and challenged

i.c. with KUNV or MVEV on day 0. In addition, two groups of 15 resistant DUB mice,

which received only a virus challenge (KUNV or MVEV) and acted as control groups

were also included. All the mice were monitored for any signs of illness for 40 days p.i.

As expected, the absence of both sets of T cells prolonged the ATD and increased

survival rate in mice infected with KUNV compared to control group mice (Figure 7.9).

Mice did not show any signs of sickness until 11 days after infection. The first deaths

(30%) were recorded 14 days after infection. Later, mortality increased to 70% on day

17 p.i. The remaining mice (30%) did not develop any signs of diseases when monitored

for up to 40 days. Interestingly, although the percentage of survival was similar to the

survival of DUB mice with depleted CD8+ T cells, the ATD was much longer in T cell-

depleted mice, suggesting that both CD4+ and CD8+ T cells acted in concert to induce

morbidity in KUNV-infected DUB mice. The ATD in T cell-depleted mice was day15.3

± 0.5 p.i. (Table 7.3), which varied considerably when compared to control mice, CD4+

T cell-depleted mice and CD8+ T cell-depleted mice (ANOVA, p < 0.007). In contrast,

the brain viral titres in T cell-depleted mice succumbing to KUNV were not

significantly different from CD4+ or CD8+ T cells depleted mice, although they were

remarkably greater than control mice (Student t test, p < 0.001).

CHAPTER 7

IMMUNOPATHOLOGY

198

0

10

20

30

40

50

60

70

80

90

100

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Days post infection

Su

rviv

al (%

)

KUNV

KUNV-CD4

KUNV-CD8

MVEV

MVEV-CD4

MVEV-CD8

Figure 7.7. Effect of CD4+ or CD8+ T cells depletion on mortality of resistant DUB

mice following challenge with KUNV and MVEV.

Fifteen mice were used in each group. Data were pooled from two separate experiments.

0

1

2

3

4

5

6

5 6 7 8 9 10 11 12 13 14

Days post infection

Lo

g1

0 T

CID

50/0

.01g

MVEV

MVEV-CD4

MVEV-CD8

Figure 7.8. Effect of CD4+ or CD8+ T cells depletion on viral titres following

MVEV infection in resistant DUB mice.

At each time point p.i., average values for virus titres were derived from 3 mice.

CHAPTER 7

IMMUNOPATHOLOGY

199

Figure 7.9. Effect of T cells (CD4+ and CD8+) depletion on mortality of KUNV and

MVEV-infected resistant DUB mice.

Fifteen mice were used in each group. Mice were infected with virus only or pre-treated

with both cytotoxic anti CD4+ and anti CD8+ monoclonal antibodies prior to virus

infection. Mice were monitored for any signs of sickness for 40 days.

0

10

20

30

40

50

60

70

80

90

100

1 3 5 7 9 11 13 15 17 19 21 23 25 27

Days post infection

Su

rviv

al (%

)

KUNV

KUNV-T cells

MVEV

MVEV-T cells

CHAPTER 7

IMMUNOPATHOLOGY

200

Table 7.3. Summary on the effect of T cells depletion in DUB mice challenged with

KUNV or MVEV.

Control CD4

depletion

CD8

depletion

CD4/CD8

depletion

KUNV

Survival (%)

0

(0/15)a

0

(0/15)

40

(6/15)

30

(3/10)

ATDb (day p.i) 9.0 1.0 10.4 0.2 12.4 0.7 15.3 0.5

Average viral

titresc (log10

TCID50 units/

0.01g)

0.6 1.0 4.6 0.7 3.3 1.4 3.8 0.8

MVEV

Survival (%)

100

(15/15)

100

(15/15)

100

(15/15)

33

(5/15)

ATDb (day p.i) - - - 22.0 2.8

Average viral

titresc (log10

TCID50 units/

0.01g)

- - - 0d

a Number of mice died/total number of mice

bAverage time to death

c Virus titres were taken from mice that succumbed to the infection

d No virus was detectable by TCID50 bioassay in mice that died from virus infection

except in one mouse which had 4 log10 TCID50 units/0.01g tissue

CHAPTER 7

IMMUNOPATHOLOGY

201

Depletion of both CD4+ and CD8+ T cells also resulted in some deaths in mice infected

with MVEV. However, disease progression was very slow. Mice were healthy and did

not show any signs of sickness until day 14 p.i., when some of them became slightly

hunched. One mouse later suffered from hind leg paralysis and was culled on day 18 p.i.

Some more deaths were recorded later; between days 20 to 27 p.i. However mice that

died between days 20 to 27 p.i. did not show typical signs of illness such as hunched

posture and ruffled fur as seen in susceptible HeJ mice i.c. infected with KUNV or

MVEV as well as resistant DUB mice that succumbed to i.c. KUNV challenge. In fact,

T cell-depleted resistant mice slowly lost weight, did not show interest in eating and

were unusually very inactive. They were eventually culled when they lost about 30% of

the normal body weight. By day 27 p.i., 10 out of 15 mice died and the rest of the mice

survived the infection when monitored for up to 40 days (67% mortality, Kaplan Meier

test, p < 0.0014 when compared to non-treated i.c. MVEV-infected mice). The ATD for

these mice was 22 ± 2.8 days p.i. (Table 7.3). No virus was detectable by TCID50

bioassay in the brains of dying resistant DUB mice except in 1 out of 10 mice (which

died on day 18 p.i.). This mouse had brain virus titres of 4 log10 TCID50/0.01g at the

time of death (Table 7.3).

On day 27 p.i., brains from two i.c. MVEV-infected DUB mice lacking T cells were

harvested and analysed for presence of T cells by flow cytometry. Interestingly,

mononuclear cells isolated from brains of these mice contained 7.22% CD4+ T cells

and 8.32% CD8+ T cells (data not shown). Exact numbers of CD4+ and CD8+ T cells

however could not be enumerated as the total number of cells from these mice was not

determined. Nevertheless, the detection of CD4+ T and CD8+ T cells in these mice at

very late of infection (day 27.p.i.) suggests that virus may be present at a low level

which was undetectable by TCID50 bioassay but inducing recruitment of inflammatory

cells into the brain.

In summary, T cell depletion (both CD4+ and CD8+ T cells) experiment provided

further insight into the role of these inflammatory cells particularly following i.c.

MVEV infection in resistant DUB mice. T cells exhibited a neuroprotection role in

resistant DUB mice during i.c. MVEV infection but the absence of both subsets of T

cells was required in order to elicit an effect on the survival of resistant DUB mice.

CHAPTER 7

IMMUNOPATHOLOGY

202

7.2.3 ANALYSIS OF CYTOKINE PRODUCTIONS

Cytokines are important regulatory proteins that involved in the regulation of the

immune response and inflammation. Deregulation of cytokine production has been

shown to be responsible for many pathological manifestations in viral diseases (Kunzi

and Pitha, 2000). T lymphocytes are required for cell-mediated immune responses and

antibody production. This dual role is controlled by two distinct T-helper (Th) cell

subsets that produce different sets of cytokines (Kunzi and Pitha, 2000). The cytokine

profile defined as Th1 is associated with production of TNFα, IFNγ, IL-2 and IL-12 and

leads predominantly to the cell-mediated immunity. Th2 cytokine profile is associated

with IL-4, IL-6 and IL-10 secretion and leads to the production of virus-specific

antibodies (Kunzi and Pitha, 2000). Since the previous section (Section 7.2.2.3 and

7.2.2.4) demonstrated that T cells were important determinants of the outcome of

infection, the levels of Th1 cytokines (TNFα, IFNγ and IL-2) versus Th2 cytokines

(IL-4 and IL-10) were examined to investigate whether there was a correlation between

particular cytokine profiles and the severity of infection. To achieve this, susceptible

HeJ and resistant DUB mice were challenged i.c. with either KUNV or MVEV and at

selected time points after infection, serum and brains from a minimum of three mice per

group were collected and used to determine the concentration of Th1 and Th2 cytokines,

using a commercial ELISA assay. The brains were prepared as 50% homogenates for

the ELISA assay.

In addition to Th1/Th2 cytokines, brain IFN type I levels were also measured in this

study as this cytokine is part of the earliest host defense mounted during viral infection

and it is also involved in modulating the immune response and regulating production of

other cytokines. IFN type I production was measured only in the brain since neurons are

the major target cell for neuroinvasive flaviviruses and this cytokine is usually released

by infected cells or tissues. However, the assay employed to measure IFN type I levels

was developed in-house and is based on the antiviral property of this cytokine.

CHAPTER 7

IMMUNOPATHOLOGY

203

7.2.3.1 Cytokine productions in susceptible HeJ mice

In the IFN type I bioassay, serial two-fold dilutions of brain homogenates were tested

for the presence of IFN type I on confluent L929 cells prior to EMCV infection. IFN

type I present in the brain homogenates protects L929 cells from EMCV infection and

consequently cell lysis. Brain IFN type I levels were determined on day 3 p.i. and at the

time of death following KUNV or MVEV i.c. challenge. No IFN type I was detected in

the brains of uninfected mice. Brain IFN levels on day 3 p.i. were 7000 ± 600 and 9600

± 1848 I.U./mL in HeJ mice infected with KUNV and MVEV, respectively, (data not

shown) and then increased to 7466.7 ± 2822.0 I.U./mL following i.c. KUNV infection

and 12800.0 ± 0.0 I.U./mL during i.c. MVEV infection at the time of death (Student t

test, p > 0.05, Table 7.4). The increase in IFN type I levels as the infection progressed

coincided with an increase in the brain viral titres (Section 6.2.1.1).

TNFα is a proinflammatory cytokine which is associated with the Th1 cytokine

response but it is not produced by T cells. Commercial ELISA was used to detect this

cytokine. However, due to limited reagents available, only serum and brain TNFα levels

from dying susceptible HeJ mice on were analysed. TNFα was not detected in the sera

of all susceptible HeJ mice tested. In contrast, susceptible HeJ mice i.c. infected with

KUNV had brain TNFα levels of 126.1 ± 38.9 pg/mL while those infected with MVEV

had on average 182.5 ± 17.5 pg/mL TNFα (Table 7.4).

Sera and brain homogenates were collected from susceptible HeJ mice infected with

either KUNV or MVEV on day 3 p.i. and at the time of death (days 5 and 6 p.i.,

respectively) and tested for Th1 cytokines, IFNγ and IL-2. As shown in Table 7.5, IL-2

could not be detected in either the sera or the brain of any of HeJ mice tested. IFNγ was

also not detected in the sera at both time points or in the brains 3 days after infection.

However, brains harvested from mice dying from KUNV challenge (day 5 p.i.) had on

average 612.5 ± 112.5 pg/mL IFNγ (Table 7.4). Notably, significantly greater brain

levels of the same cytokine (Student t test, p < 0.03) were found in susceptible HeJ mice

that died on day 6 after i.c. challenge with MVEV than in moribund susceptible HeJ

mice i.c. infected with KUNV at day 5 p.i. (Table 7.4). Similar to IFN type I and TNFα,

the higher levels of brain IFNγ in MVEV-infected HeJ mice coincided with the higher

replication levels of MVEV compared with KUNV (Figure 6.1).

CHAPTER 7

IMMUNOPATHOLOGY

204

Table 7.4. Cytokine levels in brains of susceptible HeJ mice at the time of death

following infection with KUNV or MVEV.

TNFα IFNαβ* IL-2 IFN* IL-4 IL-10

KUNV

(day 5 p.i)

126.1 ±

38.9

7466.7 ±

2822.0 BD

a

612.5 ±

112.5

178.4 ±

18.4 BD

MVEV

(day 6 p.i)

182.5 ±

17.5

12800.0 ±

0.0 BD

2222.0 ±

542.0

303.8 ±

103.8 BD

Cytokine levels were measured when HeJ mice succumbed to the virus infection; day 5

p.i for KUNV and day 6 p.i for MVEV. IL-2, IFN, IL-4 and IL-10 levels were also

measured in brains on day 3 p.i and in serum on day 3 and day 5/6 p.i; however the

values were below detection. The cytokines were also below detection in uninfected

mice. *The levels of brain IFNαβ and IFN in MVEV-infected mice were significantly

higher than in KUNV-infected mice (Student t test, p < 0.05).

aBelow detection

CHAPTER 7

IMMUNOPATHOLOGY

205

The levels of two major Th2 cytokines, IL-4 and IL-10 were studied in parallel to Th1

cytokine in the same mice. The levels detected were either too low or were below

detection level following challenge with both viruses in susceptible HeJ mice. IL-10

cytokine was below detection level in all samples tested. One of four mice had 50

pg/mL IL-4 in the brain, 3 days after challenge with KUNV virus while none of the

mice infected with MVEV had detectable IL-4. On day 5 p.i, following KUNV

infection, two out of three mice had detectable IL-4. The average values for this

cytokine from these two mice were 44.6 ± 4.6 pg/mL. Similarly, only two out of three

mice that had fatal MVEV infection showed detectable IL-4, although the average

values were slightly higher than that found following KUNV infection (76 ± 26 pg/mL).

Based on a strong production of IFN type I, TNFα and IFNγ following infection with

KUNV and MVEV, it can be concluded that fatal encephalitis induced by KUNV and

MVEV in HeJ mice was associated with a strong Th1 response.

7.2.3.2 Cytokine productions in resistant DUB mice

The levels of the same cytokines were also investigated in the sera and brains of

flavivirus resistant DUB mice after i.c. infection with KUNV or MVEV. The sera and

brain levels of these cytokines (except for IFN type I where only brain samples were

tested) were measured on days 3, 5, 7 and 9 p.i.

IFN type I levels following infection with both viruses showed a similar trend in

resistant DUB mice (Figure 7.10). On day 3 p.i., small amounts of IFN type I were

detected but the values increased sharply on day 5 p.i. following either KUNV or

MVEV infection. Following this, brain IFN type I started to diminish and on day 9 p.i.,

the level of this cytokine was the lowest detected throughout the course of infection.

However, while both viruses induced similar kinetics of IFN type I response, the brain

levels of this cytokine were found to be higher from day 3 to day 7 p.i. during MVEV

infection than KUNV infection. The difference in the amount of IFN type I produced in

MVEV versus KUNV-infected brains on day 3 p.i. was significant (Student t test, p <

0.03). However, on day 9 p.i., MVEV-infected resistant DUB mice had less than half

the amount of IFN type I ( 70.83 ± 16.35 I.U./mL) produced in mice challenged with

KUNV (180 ± 51.47 I.U./mL, the difference was significant, Student t test, p < 0.05).

CHAPTER 7

IMMUNOPATHOLOGY

206

The amount of IFN type I induced following infection with these two viruses seemed to

be in a direct correlation with the virus replication (Chapter 5, Figure 6.1).

TNFα was not detected in sera of any of the mice tested except on day 9 p.i. in mice

infected with MVEV (data not shown). At this time point, MVEV-infected resistant

DUB mice had on average, 57 ± 10.6 pg/mL TNFα in the sera. By contrast, brain TNFα

levels were detected in all mice throughout the infection with both viruses (Figure 7.10).

The amount of this cytokine was the highest on day 3 after infection with both viruses

and then slowly declined as the disease progressed. However, MVEV constantly

induced higher levels of production of this cytokine than KUNV, with the highest

difference detected on day 3 p.i. (Student T test, p < 0.05).

Th1 cytokines, IFNγ and IL-2 were not detected in the mouse sera when tested on days

3, 5, 7 and 9 p.i (Table 7.5). Similarly, IL-2 cytokine was not detected in the brains of

mice challenged with either KUNV or MVEV at the same time points p.i. (Table 7.6).

However, IFNγ was detected in every mouse brain tested following infection with either

of these two viruses 7 and 9 days after infection. The highest levels of IFNγ were

produced on day 7 p.i. and interestingly KUNV induced significantly higher IFNγ levels

than MVEV (3083 ± 282 pg/mL and 1034 ± 217 pg/mL following KUNV and MVEV

challenge respectively (p < 0.0003). On day 9 p.i., the production of IFNγ significantly

diminished in the brains of mice infected with both viruses.

IL-10 (Th2 cytokine) was only detected in the sera of three out of six DUB mice 9 days

after MVEV infection. IL-4 cytokine was detected in a proportion of mice infected with

both viruses. Following KUNV infection, one out of four mice had 24 pg/mL IL-4 3

days after infection. From the sera collected from seven resistant DUB mice i.c. infected

with MVEV, only two had measurable IL-4 at similar time points during infection. In

KUNV-infected mice which had detectable IL-4, the level of this cytokine increased to

85.88 ± 29 pg/mL on day 5 p.i. and then gradually decreased and became undetectable 9

days after infection. A similar trend was also demonstrated following MVEV infection.

CHAPTER 7

IMMUNOPATHOLOGY

207

A B

0

1

2

3

4

5

6

7

8

9

3 5 7 9

IFN

αβ

(IU

x 1

03/m

L)

0

50

100

150

200

250

300

3 5 7 9

TN

(p

g/m

L)

Days post infection

KUNV MVEV

Figure 7.10. Brain IFNαβ (A) and TNF (B) levels in resistant DUB mice following

infection with KUNV and MVEV.

Average values for brain cytokine levels were derived from 3 mice. Data presented as

average ± standard error. The cytokines were below detection in brains of uninfected

mice.

CHAPTER 7

IMMUNOPATHOLOGY

208

The levels of IL-4 in the brains of infected resistant mice were inconsistent. In addition,

the kinetics of this cytokine production in animals tested positive for IL-4 was parallel

to the kinetics of IL-4 production in the sera. The average amounts of IL-4 in IL-4-

positive animals increased from day 3 p.i. and reached the highest level on day 7 p.i.,

and then slowly declined to below detectable levels on day 9 p.i. following both virus

infections (Table 7.5 and Table 7.6). Thus, KUNV and MVEV induced cytokines with a

strong bias towards Th1 response in resistant DUB mice, similar to the observation in

HeJ mice.

7.2.3.3 Analysis of major IFNγ producing cells in resistant DUB mice

In the above study (Section 7.2.3.1 and 7.2.3.2), it was clearly demonstrated that IFNγ

response was the strongest compared to other cytokines examined. The presence of this

cytokine at the time when resistant DUB mice started to show signs of disease and

succumb to i.c. KUNV infection was quite intriguing. It is possible that IFNγ may have

a role in the pathogenesis of KUNV infection in DUB mice. Therefore, this study was

designed to identify the major cells that produce IFNγ in resistant DUB mice during

KUNV and MVEV infection by using flow cytometry analysis.

Previous data showed that the highest brain IFNγ levels were at day 7 p.i., and hence

this time point was selected to study different cell types that produce IFNγ. It is known

that T cells, NK cells and macrophages are capable of producing IFNγ following

appropriate stimulation. However, since NK cells were shown to have no apparent role

in the pathogenesis of flavivirus (Chambers and Diamond, 2003), NK cells were

excluded from this study. Brain mononuclear cells were harvested from resistant DUB

mice 7 days after virus challenge and then divided into three groups. Two groups were

dually stained for the cell surface marker of TcR (activation marker for T cells) and for

either CD4+ or CD8+ cell surface markers. The third group of cells was stained for

CD11b+ cell surface marker. Following this, intracellular staining for IFNγ was

performed in all groups of cells. Results were evaluated by flow cytometry and cells

were gated for Tcr+/CD4+, TcR+/CD8+ or CD11b+ prior to analysing IFNγ+ cells. The

actual number of cells positive for IFNγ was calculated from the total numbers of

CD4+, CD8+ or CD11b+ cells as the percentage of IFNγ+ cells (obtained from flow

cytometric analysis).

CHAPTER 7

IMMUNOPATHOLOGY

209

As shown in Figure 7.11A, CD11b+ cells were the predominant cell type found in the

brains of KUNV and MVEV-infected DUB mice. The second largest population was

CD8+ T cells and these data were in agreement with the previous quantitative analysis

performed on brain mononuclear cells of infected resistant mice (Section 7.2.1.2).

However, on day 7 p.i. after KUNV and MVEV infection, CD8+ T cells were the main

producers of IFNγ (Figure 7.11B). The numbers of CD8+ T cells that produced IFNγ

were more than 4 times greater than the numbers of IFNγ-producing CD4+ or CD11b+

cells (Figure 7.11B). KUNV infection on average induced slightly higher numbers of

IFNγ+ CD8+ T cells (4.58 ± 0.13 x 103 cells) than MVEV infection (4.13 ± 0.1 x 10

3

cells). The numbers of CD4+ T cells that produced IFNγ were lower; 1.38 ± 0.4 x 103

and 1.76 ± 0.13 x 103 cells in the brains of DUB mice challenged with KUNV and

MVEV, respectively. Interestingly, although CD11b+ cells were the predominant cell

type found in the brain, they were less efficient producer of IFNγ compared to T cells.

However, 1.305 ± 0.37 x 103 CD11b+ cells produced IFNγ in KUNV-infected mouse

brains, which was significantly higher (p<0.03) than that found after MVEV infection

(0.37 ± 0.07 x 103 cells). The average of total IFNγ producing cells were 7.2 x 10

3 and

6.2 x 103 cells found in DUB mice following i.c. KUNV and MVEV challenge,

respectively.

In summary, the data presented here showed that CD8+ T cells were the major

producers of IFNγ in the brains of resistant mice following i.c. infection with either

KUNV or MVEV. This finding bears a great significance and in combination with the

data presented in the previous sections highlights the role of CD8+ T cells in KUNV

pathogenesis in the model of genetically resistant mice. In addition, this finding also

suggests that CD8+ T cells may exert detrimental effect in infected resistant DUB mice

through IFNγ production.

CHAPTER 7

IMMUNOPATHOLOGY

210

Table 7.5. Th1-Th2 cytokines in DUB mouse sera following infection with KUNV

and MVEV.

IFNγ

(pg/mL)

IL-2

(pg/mL)

IL-4

(pg/mL)

IL-10

(pg/mL)

KUNV

d3

-a

-

-

-

-

-

-

-

-

-

24.0

-

-

-

-

-

d5

-

-

-

-

-

-

156.2

156.2

50.0

43.0

-

24.0

-

-

-

d7

-

-

-

-

-

-

-

-

-

-

-

-

85.8

106.8

91.5

61.2

34.5

52.3

33.2

42.2

-

-

-

-

-

-

-

d9

-

-

-

-

-

-

-

-

-

-

-

-

MVEV

d3

-

-

-

-

-

-

-

-

37.8

48.0

-

-

-

-

-

-

-

-

d5

-

-

-

-

-

-

156.2

109.34

-

-

-

-

-

-

-

d7

-

-

-

-

-

-

-

-

-

-

-

-

-

134.0

230.0

134.0

-

23.4

24.0

-

-

-

98.4

98.4

70.4

d9

-

-

-

-

-

-

-

-

-

-

-

-

The cytokines were detected by ELISA and assay sensitivities were 2000-15pg/mL,

200-2pg/mL, 500-4pg/mL and 2000-15pg/mL for IFNγ, IL-2, IL-4 and IL-10,

respectively. The cytokines were also below detection in uninfected mice. Each number

represent sample taken from one mouse. aBelow detection.

CHAPTER 7

IMMUNOPATHOLOGY

211

Table 7.6. Th1-Th2 cytokines in DUB mouse brains following infection with KUNV

and MVEV.

IFNγ*

(pg/mL)

IL-2

(pg/mL)

IL-4

(pg/mL)

IL-10

(pg/mL)

KUNV

d3

- a

-

-

-

-

-

-

-

30.0

-

-

-

-

-

-

-

d5

-

-

-

-

-

-

190.0

169.2

-

68.0

185.0

-

-

-

d7

2880.0

3706.0

2823.0

3278.0

3857.0

1957.0

-

-

-

-

-

-

-

171.0

177.0

-

-

-

55.0

160.0

-

-

-

-

-

-

d9

432.0

458.0

408.0

-

-

-

-

-

-

-

-

-

MVEV

d3

-

-

-

-

-

-

-

-

95.0

-

50.0

30.0

-

-

-

-

d5

-

-

-

-

-

-

105.0

200.0

240.0

-

-

-

d7

632.0

662.0

706.0

1280.0

927.0

2000.0

-

-

-

-

-

-

-

-

-

134.0

230.0

134.0

70.0

105.0

68.0

-

-

-

-

-

-

d9

233.7

433.0

400.0

-

-

-

-

-

-

-

-

-

The cytokines were detected by ELISA and assay sensitivities were 2000-15pg/mL,

200-2pg/mL, 500-4pg/mL and 2000-15pg/mL for IFNγ, IL-2, IL-4 and IL-10,

respectively. The cytokines were also below detection in uninfected mice. Each number

represent sample taken from one mouse. aBelow detection. *IFNγ levels were

statistically different during KUNV versus MVEV infection on day 9 p.i.

CHAPTER 7

IMMUNOPATHOLOGY

212

A

Total number of cells

0

50

100

150

200

250

300

350

400

450

500

CD8+TcR+ CD4+TcR+ CD11b+

Nu

mb

er

of

cells (

x 1

03)

B

IFNγ producing cells

0

1

2

3

4

5

6

CD8+TcR+ CD4+TcR+ CD11b+

Nu

mb

er

of

cell

s (

x10

3)

KUNV MVEV

Figure 7.11. Analysis of IFN producing cells in resistant DUB mouse brains 7 days

after infection with KUNV or MVEV.

(A) Total numbers of brain CD8+ T cells, CD4+ T cells and microglia/macrophages

(CD11b+) isolated from KUNV or MVEV-infected DUB mice (B) Numbers of CD8+ T

cells, CD4+ T cells and microglia/macrophages that produced IFNγ. *Significant

different observed in KUNV induced CD11b+ cells verus MVEV induced CD11b+

cells (Student t test, p < 0.03)

*

CHAPTER 7

IMMUNOPATHOLOGY

213

7.3 DISCUSSION

Disease severity is a function of either a direct virus-induced cytopathology and/or a

virus triggered excessive or inappropriate immune response (Brehm et al, 2004). In the

previous chapter (Chapter 6), histological analysis of brain tissue sections harvested

from KUNV and MVEV-infected susceptible and resistant mice suggested that the

extent of brain inflammation is in a direct correlation with the virus strain but not with

the outcome of disease. However, this information is rather general and in order to

obtain a better insight into the critical factors that dictate the severity of infection, a

more detailed analysis targeting specific mononuclear cell subtypes recruited into the

infected brains was carried out. In this chapter, a more sensitive approach utilising flow

cytometry was used to quantify four types of inflammatory cells, CD4+ T cells, CD8+ T

cells, B cells and CD11b+ cells.

It was shown in this study (Section 7.2.1.2) that i.c. MVEV challenge resulted in greater

numbers of inflammatory cells (in total) recruited into the brains of infected susceptible

HeJ and resistant DUB mice than KUNV infection. This was in agreement with the

histopathological studies as described in the preceding chapter. However, interestingly,

the quantities of specific mononuclear cell subtypes recruited to the mouse brains were

different during different flavivirus infections. While KUNV caused more infiltration of

CD8+ T cells to the brains of susceptible HeJ and resistant DUB mice, MVEV induced

greater recruitment of CD4+ T cells, B cells as well as CD11b+ cells.

Although greater brain tissue inflammation was observed and more brain mononuclear

cells were isolated from KUNV and MVEV-infected susceptible HeJ mice at the time of

death than from infected resistant DUB mice from day 6 to day 9 p.i., the numbers of

brain CD4+, CD8+ and B cells were at least 10 times lesser in susceptible HeJ mice.

More importantly, T cells, B cells and CD11b+ cells that were isolated from the brains

of susceptible HeJ mice represented only approximately half of the total cells infiltrating

the brains of HeJ mice. The identity of the remaining infiltrating cells in HeJ mouse

brains is unknown and should be further investigated. However, the possibility is that

NK and polymorphonuclear cells may accumulate in the brains of susceptible HeJ mice

more readily than in resistant DUB mice during KUNV and MVEV infections. Brain

NK cells were shown to be pathogenic during virulent Semliki Forest infection in

C57BL/6J mice (Alsharifi et al, 2006).

CHAPTER 7

IMMUNOPATHOLOGY

214

A considerable numbers of research have been conducted recently to elucidate the role

of CD8+ T cells in flavivirus infection of susceptible hosts (reviewed in Chambers and

Diamond, 2003; An et al, 2004). Because of this, functions and implications of CD8+ T

cells in flavivirus infections are better understood than CD4+ T cells. CD8+ T cells

were shown to have a neuroprotective role during infection with WNV New York strain

and low doses of WNV Sarafend (Shrestha and Diamond, 2004; Wang et al, 2003b). In

contrast, CD8+T cells contribute to the pathogenesis of WNV Sarafend when mice were

infected at high doses (Wang et al, 2003b). In this study it was found that the greater

numbers of CD8+ T cells present in KUNV-infected susceptible mice compared to that

observed in MVEV-infected HeJ mice did not contribute to either protection or

pathogenesis of KUNV, as determined by the CD8+ T cells depletion study (Figure 7.6

and Table 7.3). CD8+ T cell-depleted susceptible HeJ mice had similar rate of

susceptibility to i.c. KUNV infection as control susceptible HeJ mice. Thus, this

suggests that susceptible HeJ mice succumb to i.c. KUNV infection because of a direct

virus infection with unrestricted replication in the CNS and CD8+T cells seem not to

contribute to the severity of KUNV infection. In contrast, absence of CD8+ T cells

rendered MVEV-infected HeJ mice more susceptible to the infection, demonstrated by

the shorter ATD (one day earlier). Since T cells have no significant

immunopathological role in susceptible HeJ mice, it would be interesting in future to

determine other inflammatory cells that could contribute to the fatal encephalitis caused

by KUNV and MVEV.

Although a slight protective role of CD8+ T cells against MVEV infection in HeJ mice

was in agreement with the role of CD8+ T cells in the host defence against WNV NY

strain and low doses of WNV Sarafend infection (Shrestha and Diamond, 2004; Wang

et al, 2003b), the mechanism by which CD8+ T cells contribute to the control of disease

severity following MVEV infection in susceptible HeJ mice is unknown. Contrary to

the previous reports which suggest that CD8+ T cells play essential role in controlling

and clearing flavivirus infection (Shrestha and Diamond, 2004; Wang et al, 2003b), in

this current study, the lack of CD8+ T cells did not affect brain virus titres in

susceptible HeJ mice.

Virus-specific CD4+ T cells have disparate roles in flavivirus infection, depending on

the model of infection used. For instance, the lack of CD4+ T cells does not increase

CHAPTER 7

IMMUNOPATHOLOGY

215

susceptibility of mice to DENV infection (Shresta et al, 2004). In contrast, CD4+ T

cells are important for priming memory CD8+ T cells during YFV infection in mice

(Liu and Chambers, 2001). Recently, CD4+ T cells have been suggested to have

immunoprotective role by sustaining antibody production and effector CD8+ T cells in

the CNS during the infection with WNY NY strain in susceptible C57/BL6 mice (Sitati

and Diamond, 2006). The absence of CD4+ T cells resulted in persistent WNV infection

in the brain and consequently 100% mortality of infected susceptible C57/BL6 mice

while only 30% of control C57/BL6 mice died from WNV infection (Sitati and

Diamond, 2006). In contrast, data presented in this chapter suggest a slight detrimental

effect of CD4+ T cells in susceptible HeJ mice following i.c. challenge with KUNV and

MVEV. Twenty percent of KUNV or MVEV-infected HeJ mice lacking CD4+ T cells

had a delayed death (a day later compared to control mice) (Figure 7.6 and Table 7.3).

However, since only a small numbers of susceptible HeJ mice involved in CD4+ T cell

depletion experiment (5 mice), this study should be repeated in future to allow statistical

analysis of the experiment.

Previously published studies have demonstrated that resistant and susceptible mice

mount immune responses with similar kinetics and extent (reviewed in Brinton and

Perelygin, 2003). As the flavivirus resistance gene only restricts virus replication and

spread, an intact immune response is usually required to clear the virus from an infected

host. If the virus is not cleared, resistant mice generally will die from the infection

(reviewed in Brinton and Perelygin, 2003). However, this conclusion was derived from

a limited number of studies using selected flaviviruses in resistant RV mice (Goodman

and Koprowski, 1962a; Bhatt and Jacoby, 1976). It was shown that immunosuppression

of i.p. Banzi-infected RV mice with treatments such as x-irradiation, cyclophosphamide

or thymectomy decreased their survival time (Bhatt and Jacoby, 1976). Interestingly,

cyclophosphamide did not change the ATD in i.c. Banzi-infected resistant RV mice,

suggesting that inoculation route may influence the beneficial or detrimental role of the

host antiviral reaction. Unfortunately, no further studies have been performed to look at

the role of specific immune cells in preventing or contributing to fatal encephalitis in

flavivirus resistant mice.

Quantitative analyses showed that MVEV induced almost similar numbers of CD4+ and

CD8+ T cells throughout the course of infection in resistant DUB mice (except on day 5

p.i.). In contrast, CD8+ T cells were about double or triple the numbers of CD4+ T cells

CHAPTER 7

IMMUNOPATHOLOGY

216

following KUNV challenge (from day 7 to 9 p.i.) (Figure 7.3). Contributing factors that

lead to the different polarisation of recruited lymphocytes are unknown and requires

further studies. Chemokines and cytokines are known to be crucial for signalling and

recruiting lymphocytes to the infected areas (Kunzi and Pitha, 2000). Neuronal

CXCL10 chemokine for instance, directs CD8+ T cell recruitment to the brain during

WNV infection (Klein et al, 2005). In this current study, the levels of proinflammatory

cytokines during i.c. KUNV versus MVEV infection in resistant DUB mice were

different (to be discussed later) but the beneficial or damaging effect of these cytokines

and their involvement in recruiting peripheral inflammatory cells are yet to be

determined. Additionally, while the numbers of CD4+ T cells increased over time

following both virus infections, CD8+ T cell numbers decreased after MVEV infection

but did not vary significantly during the course of KUNV infection in resistant DUB

mice (except on day 5 p.i.). Activated T cells migrating to the brain parenchyma usually

are not retained in the CNS unless contact with antigen is maintained (Kimura and

Griffin, 2003). Thus, the possibility exists that the low levels of infectious virus or

virus antigen were responsible for attracting and sustaining these inflammatory cells,

particularly CD8+ T cells in the brains of resistant DUB mice 9 days after KUNV

infection.

The decline in the total numbers of splenocytes during the course of KUNV and MVEV

infection in resistant DUB mice coincided with the increased numbers of lymphocytes

in the brain (Figure 7.1). This strongly implicated cell migration from the spleens to the

brains, as opposed to extensive local cell division within the CNS, as the major source

of lymphocytes accumulation in the brain. Intriguingly, as observed in the brains, there

were higher numbers of splenocytes isolated from MVEV-infected resistant DUB mice

than from KUNV-infected mice. The reason for such occurrence is unknown although it

is possible that KUNV may induce apoptosis of splenocytes in infected resistant DUB

mice.

The prevalence of T cells in the brain particularly late in infection usually is an

indication of their role in the recovery or severity of diseases in flavivirus infection. In

this study, KUNV induced-T cells have been demonstrated for the first time to have an

immunopathological role and to be involved in the fatal disease of resistant DUB mice.

More importantly, CD8+ T cells were shown to play a larger part on the development of

lethal encephalitis than CD4+ T cells during i.c. KUNV infection in resistant DUB

CHAPTER 7

IMMUNOPATHOLOGY

217

mice, probably due to the greater numbers of CD8+ T cells recruited into the brain than

CD4+ T cells (Figure 7.6). This was apparent by the delayed ATD as well as higher

survival rate of CD8+-depleted resistant DUB mice than control DUB mice or CD4+-

depleted DUB mice (Table 7.3). It is worth noting here that although prolonged ATD

was observed in KUNV-infected resistant DUB mice lacking CD4+ or CD8+ T cells,

these mice had significantly greater brain viral burdens at the time of death than dying

control resistant DUB mice infected with KUNV only. This suggests that a) cellular

immunity is important in clearing flavivirus infection, which is in agreement with other

studies (Wang et al, 2003b; Shrestha and Diamond, 2004) and b) high viral titres alone

in the absence of CD4+ and CD8+ T cells could not promote fatal disease in KUNV-

infected resistant mice.

Another important finding of this study was that depletion of both CD4+ and CD8+ T

cells promoted longer survival time of resistant DUB mice than a transient loss of a

single subset of T cells during i.c. KUNV challenge (Figure 7.9). This suggests that

immunopathological role of CD4+ and CD8+ T cells were not redundant during i.c.

KUNV infection in resistant DUB mice as depletion of total T cells had a synergistic

effect on the average survival time. However, T cell depletion could not prevent

complete death of KUNV-infected resistant DUB mice as 70% of mice lacking both T

cells subsets eventually succumbed to the infection, indicating that there were other

factors involved in the pathogenesis of KUNV.

Unexpected yet interesting findings were obtained during i.c. MVEV challenge in

resistant DUB mice lacking either CD4+ or CD8+ T cells. Depletion of either CD4+ or

CD8+ T cells alone did not render these animals susceptible to MVEV infection at all

(Figure 7.7). This suggests that the lack of either CD4+ or CD8+ T cells was not critical

for neuroprotection of DUB mice against MVEV.

Interestingly, when infectious MVEV was cleared from the brains of control infected

resistant DUB mice on day 9 p.i., CD4+ T cell-depleted as well as CD8+ T cell-

depleted DUB mice still had relatively high brain viral titres despite the absence of any

sign of sickness (Figure 7.8). Similar to that observed during i.c. KUNV challenge,

these results further implicate the presence of additional factors, in addition to virus

titres, that are required to promote fatal disease in resistant DUB mice following i.c.

flavivirus challenge. Mice depleted of CD4+ and CD8+ T cells eventually cleared

CHAPTER 7

IMMUNOPATHOLOGY

218

MVEV in the brain on day 14 p.i. by which time these cells repopulated the brain.

Alternatively, following the absence of one T cell subset (such as CD4+ T cells), the

other subset of T cells (such as CD8+ T cells) together with the Flvr-like gene probably

acted in concert to reduce and eventually clear MVEV from the infected brains although

not as efficiently as when both subsets of T cells were present.

In contrast, the absence of both CD4+ and CD8+ T cells (all T cells) resulted in resistant

DUB mice to be highly susceptible to i.c. MVEV challenge (Figure 7.9) (67% mortality

rate). On average, the death of these mice occurred on day 22 p.i., which was 7 days

longer than KUNV-infected DUB mice that received similar treatment. Although only

one out of ten sick MVEV-infected DUB mice had a detectable brain virus titre, it is

possible that all resistant DUB mice that succumbed to MVEV infection following

depletion of T cells still had a low presence of infectious virus which was undetectable

by TCID50 bioassay. Alternatively, virus antigen rather than the infectious virus may

present in the brains of these mice. This was supported by the analysis of mononuclear

cells isolated from two mice that died on day 27 p.i. In these mice, CD4+ and CD8+ T

cells were still detected at the time of death. The recruitment and maintenance of

peripheral immune cells in the brain parenchyma occurs typically in the presence of

virus since T cells are rarely retained in the brain unless they are in contact with the

virus antigen. Due to the lack of T cells to clear the virus early, it is possible that MVEV

was retained for a prolonged period of time and this may have caused some functional

damage to the CNS cells especially the neurons. When the effect of depleting antibodies

diminished and T cells re-entered the brain, the host adaptive immune response might

be too late to prevent fatal disease outcome. In fact, T cells recruited at a very late stage

in infection may be immunopathogenic and could exacerbate the course of MVEV

infection in T cell-depleted resistant DUB mice. Signs of diseases displayed in T cell-

depleted resistant DUB mice following MVEV infection were different to those

typically shown by susceptible HeJ mice suffering from flavivirus-induced encephalitis.

The lack of interest in feeding, lethargy and weight lost also further suggests that in the

sick resistant DUB mice, normal neuronal functions in certain parts of the brain were

affected.

Experimental work conducted in this chapter put an emphasis on the role of T cells

during the challenge with two flaviviruses. However, it should be mentioned here that

brain CD11b+ cells were the predominant mononuclear cells found during MVEV and

CHAPTER 7

IMMUNOPATHOLOGY

219

KUNV challenge in both strains of mouse. The numbers of these cells were about 10

times more than the other identified leucocytes. Future studies perhaps should be

carried out to further characterise CD11b+ cell population during different flavivirus

infections in different mouse strains since various cells including microglia,

macrophages and neutrophils are known to express CD11b+ cell surface markers.

Resting microglia and activated microglia/recruited macrophages can be distinguished

by the level of expression of cell surface marker CD45 (Sedgwick et al, 1991). While

resting microglia express CD45low

/CD11b+, activated microglia and recruited

macrophage can be readily identified by CD45high

/CD11b+ surface marker expression

(Ford et al, 1995). In addition, dual staining of CD11b+ and GR1 cell surface markers

would help to identify neutrophils. These cells have been implicated in the pathogenesis

of MVEV strain 3749 in weanling Swiss mice since depletion of neutrophils coincides

with prolonged survival time and decreased mortality (Andrews et al, 1999). TNFα was

thought to be responsible for triggering the production of neutrophil-attracting

chemokines in these mice. Since TNFα was detected in infected HeJ and DUB mice

(see later), neutrophils could be present in the brains of HeJ and DUB mice during

KUNV or MVEV infection.

Development of lethal encephalitis in susceptible HeJ mice during KUNV and MVEV

infection coincided with high production of Th1 and proinflammatory cytokines

(Section 7.2.2.2). However, MVEV induced greater levels of IFN type I, TNFα and

IFNγ than KUNV in the brains of susceptible HeJ mice at the time of death, probably

due to the greater levels of brain MVEV titres in these mice. TNFα can induce

expression of adhesion molecules and chemokine synthesis in cerebrovascular

endothelial cells and astrocytes. This in turn will facilitate leucocyte extravasation and

recruitment in the CNS (Aloise, 2001). In agreement with this, as shown in Chapter 5,

stronger brain inflammation was observed in MVEV-infected HeJ mice compared to

KUNV-infected mice. However, the reason for greater levels of IFNγ during i.c. MVEV

challenge in susceptible HeJ mice is unknown, given that less brain CD8+ T cells were

detected in these mice compared to KUNV-infected HeJ mice.

Following i.c. inoculation of KUNV and MVEV in resistant DUB mice, an elevated

production of IFN type I and TNFα were detected early in the infection and then they

gradually reduced over time, which paralleled virus reduction or clearance (Section

7.2.3.1). The secretion of TNFα as early as day 3 p.i., in advance of any histologic

CHAPTER 7

IMMUNOPATHOLOGY

220

evidence of brain inflammation suggested that intrinsic CNS cells particularly microglia

were the main producers of TNFα. Furthermore, astrocytes and neurons have been

reported to be capable of producing TNFα (Munoz-Fernandez and Fresno, 1998).

Similar to that observed in HeJ mice, greater levels of brain IFN type I and TNFα

productions were seen in MVEV-infected DUB mice which may be responsible for the

more vigorous brain tissue inflammation demonstrated in DUB mice infected with this

virus than KUNV. In contrast, KUNV infection induced higher IFNγ production in

resistant DUB mice (Section 7.2.3.2). The detection of IFNγ in the brain tissue but not

in the sera indicated that effector function of T cells was only acquired in the brain (at

the site of infection within the CNS). The greatest IFNγ levels were observed on day 7

p.i. when KUNV-infected DUB mice started to show signs of diseases and production

of this cytokine reduced dramatically on day 9 p.i. when mice succumbed to the

infection.

CD8+ T cells were demonstrated to be the major IFNγ producing cells on day 7 p.i.

during i.c. KUNV and MVEV challenge in resistant DUB mice (Section 7.2.3.3). CD4+

and CD11b+ cells were also capable of producing IFNγ, albeit less efficient. This could

be an indication of the immunopathogenic role of this cytokine. It is possible that T

cells, particularly CD8+ T cells, participated in the development of lethal KUNV

infection through IFNγ production. However, further work using IFNγ neutralising

antibodies or resistant DUB mice deficient in functional IFNγ during KUNV or MVEV

i.c. challenge could confirm the function of T cells in resistant DUB mice.

In addition to IFNγ, another effector function of CD8+ T cells is cytolysis. Killing of

the target cells by Tc cells occurs primarily via the granule exocytosis pathway or the

Fas-Fas ligand mechanism. Antigen-specific killing by CD8+ T cells requires the

migration of lymphocytes to the site of infection. Shrestha and Diamond (2004) showed

that killing of target cells occurs by perforins during infection with WNV NY strain. In

contrast, during WNV Sarafend infection, the survival of mice is partially dependent on

exocytosis and/or Fas-mediated cytolytic activity (Wang et al, 2004b). However, both

findings strongly suggested that the virus clearance following flavivirus infection partly

contributed by the cytolytic mechanism of T cells. Initial study conducted in our lab

(data not shown) indicates that MVEV infection in resistant mice induced earlier

cytolytic activity compared to KUNV infection. This may contribute to early clearance

of infectious MVEV in resistant DUB mice.

CHAPTER 7

IMMUNOPATHOLOGY

221

The MHC molecule expression on CD11b+ cells from infected DUB mice was

examined from day 5 p.i. onwards (Section 7.2.1.3). The up-regulation of this cell

surface molecule can be induced by IFN type I, IFNγ as well as TNFα. Early in

infection, MHC molecule expression is induced by IFNαβ while later in the infection,

expression of these molecules is maintained by IFNγ (Kunzi and Pitha, 2000). In

addition, flaviviruses are capable of inducing expression of MHC class I molecules

independent of cytokines (King et al, 2004). Since levels of these cytokines and brain

viral titres were different in DUB infected with KUNV and MVEV, MHC molecule

expression in these mice was expected to be different. However, it is not clear why

CD11b+ cells isolated from MVEV-infected DUB on day 5 p.i. had a minimal MHC I

expression, since the levels of brain proinflammatory cytokines (IFNαβ and TNFα) and

viral titres were higher in these animals than in KUNV-infected DUB mice.

Unfortunately, the data were generated from a single experiment and needs to be

repeated. However, the greater levels of MHC molecules expression on CD11b+ cells

from KUNV-infected DUB mice than MVEV-infected DUB mice seemed to directly

correlate with the higher numbers of CD8+ T cells recruited to the brains of KUNV-

infected DUB mice.

The level of MHC I up-regulation in infected resistant DUB mice was initially

monitored in parallel with the immunostaining of viral antigens experiment.

Unfortunately, the latter analysis aimed at investigating the presence of viral antigen in

brain macrophages/microglia failed due to technical problems. Although previous

studies indicated that neurons are the preferential site of replication for flaviviruses and

that microglia could not be infected in vitro, this does not exclusively preclude the

ability of flavivirus to infect CD11b+ cells at a very low level.

In conclusion, the data presented in this chapter indicate that i.c. infection with KUNV

or MVEV induced recruitment of different types of host immune cells that may result in

the engagement/activation of different pathways of pathogenesis/viral clearance of these

flaviviruses in susceptible HeJ and resistant DUB mice. Several major findings of the

study described in this chapter were: 1) KUNV induced greater recruitment of CD8+ T

cells into the brains of infected susceptible and resistant mice than MVEV, which

coincided with the development of severe infection 2) CD8+ T cells had

immunoprotective role in MVEV-infected HeJ mice since the absence of CD8+ T cells

CHAPTER 7

IMMUNOPATHOLOGY

222

induced earlier deaths in these mice 3) However, direct CNS viral replication was more

likely to have a larger contribution to fatal encephalitis in susceptible HeJ mice

following i.c. KUNV or MVEV challenge than T cell accumulation 4) CD8+ T cells

had a greater contribution than CD4+ T cells to KUNV-induced immunopathological

disease in resistant mice 5) T cells were pivotal in flavivirus clearance from brains of

infected resistant mice although the presence of high viral titres did not always coincide

with the fatal disease outcome 6) KUNV induced a significantly greater production of

brain IFNγ than MVEV in resistant mice at later time p.i. and this cytokine was

produced predominantly by CD8+ T cells, highlighting the immunopathological role of

CD8+ T cells in KUNV infection. Combined, these findings confirmed the dual role of

T cells particularly CD8+ T cells in flavivirus infection, depending on various factors

including the virus and mouse strain used (Wang et al, 2003b; Shrestha and Diamond,

2004). It remains to be elucidated what other elements of the host antiviral immunity

that could also have a role in the death of these mice.

CHAPTER 8

GENERAL DISCUSSION

223

8.0 CHAPTER 8: GENERAL DISCUSSION

The ratio of apparent to inapparent infections following flavivirus infection in humans is

quite low, suggesting that the complex interaction between the virus and the host that

could result in a poor outcome of infection only occurs in a small number of cases

(reviewed in Chambers and Diamond, 2003). In this study, 3 flaviviruses, KUNV,

MVEV and WNV and two congenic mouse strains, flavivirus susceptible HeJ and

resistant DUB mice were used to study these interactions that could act in many discrete

ways to produce different outcomes of infection or severity of diseases (Solomon and

Mallewa, 2001).

Laboratory mice were shown to develop encephalitis following flavivirus infection,

similar to that observed in humans (Sabin, 1954; reviewed in Urosevic and Shellam,

2002). Because of this, they have been used as a small animal model to study the

pathogenesis of flavivirus-induced encephalitis. Most laboratory mouse strains are

susceptible to flavivirus infections whereas resistance is prevalent in wild type mice

(reviewed in Urosevic and Shellam, 2001). This resistance is conferred by a single

autosomal dominant gene which is known as Flv (Sabin, 1952b; Green, 1989). To study

the mechanism of resistance, several congenic mouse strains have been developed by

introducing resistance genes from wild mice into the genetic background of susceptible

HeJ mice (Sangster et al, 1993; Urosevic et al, 1999). Two strains of resistant mice,

MOLD and DUB were developed in this laboratory and they were shown to express

different degrees of resistance to flavivirus infection (Urosevic et al, 1999).

Protection conferred by the flavivirus resistance gene is known to be incomplete since

several flaviviruses have been shown to induce fatal encephalitis in flavivirus resistant

mice when infected by the i.c. route (Shueb et al, 2005; Sabin, 1954; reviewed by

Brinton and Perelygin, 2003). However, the cause of death of these resistant mice has

never been elucidated. This project was designed to identify molecular and cellular

factors responsible for the development of fatal disease in resistant mice following i.c.

infection with KUNV. In addition, the pathogenicity of KUNV was compared to the

pathogenicity of two other closely related flaviviruses, MVEV and WNV in susceptible

HeJ and resistant DUB mice during i.p. and i.c. challenge.

CHAPTER 8

GENERAL DISCUSSION

224

In the early part of this study, in vivo and in vitro characterisation of KUNV, MVEV

and WNV was carried out. As shown in Chapter 4, KUNV was non-neuroinvasive in

adult susceptible HeJ and resistant DUB mice although it could induce high morbidity

in young HeJ mice during i.p. infection. In contrast, a high mortality was observed

following i.c. inoculation in both strains of mice and in fact, KUNV was the most

neurovirulent virus compared with WNV to resistant DUB mice. Meanwhile, WNV

displayed neuroinvasiveness only in HeJ mice but was neurovirulent to both strains of

mice tested. Interestingly, i.p. and i.c. MVEV challenge resulted in high mortality of

susceptible HeJ mice. However, MVEV did not induced severe disease in DUB mice

during i.c. infection. Combined with data from in vitro characterisation of similar

viruses (to be discussed later), it was demonstrated here that each of the flaviviruses

used in this study ‘favoured’ a different mouse strain or model of infection, which is a

remarkable finding. While MVEV is highly virulent to susceptible HeJ mice, KUNV

and WNV display higher virulence/tropism for DUB mice and cell cultures,

respectively.

KUNV was not neuroinvasive in adult HeJ mice and it did not induce death during i.p.

inoculation despite various treatments used to modulate macrophage response or alter

BBB permeability in adult mice. Some investigators have suggested that a glycosylation

of the E protein is an important determinant of the flavivirus neuroinvasiveness (Shirato

et al, 2006). The exact mechanism related to the E protein glycosylation-associated

neuroinvasion is not known but it may be due to the increased binding of virus to the

cell receptors and its greater penetration into the cells resulting in enhanced viral

infectivity (Chambers and Diamond, 2003). Thus, possibly, KUNV may be unable to

replicate efficiently in peripheral tissues compared with WNV, leading to the lack of

neuroinvasiveness of this virus. Alternatively, the E protein glycosylation may increase

virus virulence indirectly, by enhancing the vulnerability of the infected host as

suggested by Shirato and co-workers (2006). These investigators have recently

demonstrated that a glycosylated variant of WNV induced higher serum TNFα levels

than non-glycosylated virus (Shirato et al, 2006) and this cytokine has been linked to

BBB breaching leading to early viral invasion into the brain (Wang et al, 2004a).

KUNV MRM16 was previously shown to be a non-glycosylated virus while WNV

Sarafend carries a glycosylated E protein (Scherret et al, 2001). However, the

glycosylation status of the same viruses used in the project is unknown since the number

CHAPTER 8

GENERAL DISCUSSION

225

of laboratory passages could affect this. It remains to be investigated whether the

observed different neuroinvasiveness of KUNV and WNV is caused by this feature.

In addition to innate resistance to flaviviruses, other host factors could also contribute to

the severity of disease following virus infection. One of the most crucial steps leading to

fatal encephalitis is the virus invasion of the brain. The mechanisms that allow

flaviviruses to enter the brain involve either breaching the BBB or evading it. Since

breaching of the BBB is considered the major mechanism of viral entry into the brain,

transient change in the BBB permeability after SDS treatment was examined in mice.

As shown in Chapter 4, SDS treatment rendered adult HeJ mice more susceptible to i.p.

WNV and MVEV infection although this was only observed when SDS was given 2 or

3 days after infection. In addition, this treatment also exacerbated the course of KUNV

infection in young HeJ mice.

The activation of macrophages is a critical step in the early non-specific defense against

many viral infections (Guidotti and Chisari, 2001) and therefore, these cells represent a

very important factor in determining the outcome of infection. Studies have shown that

the absence of macrophages renders animals highly susceptible to viral infections (Ben-

Nathan, 1996). Accordingly, a single injection of liposome encapsulated clodronate

aimed at transient depletion of macrophages very early in the infection (but not

throughout the course of infection) resulted in increased mortality rate and rapid ATD in

HeJ mice during i.p. WNV infection (Chapter 4). This suggested that the initial control

of WNV replication by macrophages is crucial for reducing severity of infection. The

lack of macrophages may allow early viraemia and consequently early virus invasion of

the CNS.

It was shown in this study that macrophages could also be pathogenic in HeJ mice

during i.p. WNV challenge, thus further supporting earlier findings in this laboratory

regarding the dual roles of macrophages during certain flavivirus infections (Pantelic,

2004). Monocytes/macrophages are known to be the principal site of replication for

DENV and they have important roles in DENV pathogenesis through the antibody-

dependent enhancement of infection and production of cytokines/chemokines (Chen and

Wang, 2002; Cardosa et al, 1986; Gollins and Porterfield, 1984). With other

flaviviruses, it is yet to be determined which peripheral tissues or organs are important

sites of viral replication, and whether the magnitude of viral production in these tissues

CHAPTER 8

GENERAL DISCUSSION

226

determines viral invasion of the CNS (reviewed in Chambers and Diamond, 2003). In

vitro and in vivo studies in this project indicated that macrophages could support WNV

replication and consequently, these cells could contribute to the pathogenesis of this

flavivirus in HeJ mice (Chapter 4 and Chapter 5). Thioglycollate injection significantly

increased the availability of permissive cells in the peritoneum (peritoneal

macrophages) (Pantelic, 2004) and consequently, exacerbated the severity of disease in

i.p. WNV infected-HeJ mice, as shown by the 100% death and shorter survival time

compared with control mice (Chapter 4). When thioglycollate-elicited macrophages

were isolated and cultured for in vitro characterisation of WNV, KUNV and MVEV, it

was demonstrated that WNV has a better infectibility in these cells than the other two

flaviviruses (Chapter 5). This was evidenced by the higher WNV titres in thioglycollate-

elicited macrophage cultures. In addition, in vitro WNV infection of these cells elicited

higher levels of TNFα production three days after infection than KUNV and MVEV

infection. This proinflammatory cytokine, as previously mentioned, can facilitate viral

invasion of the CNS by increasing the BBB permeability (Wang et al, 2004a). Similar

high infectibility of WNV has also been shown during in vitro infection in monocytes

and monocyte-derived macrophages may be an indication that monocytes/macrophages

could also be infected in vivo and be transmitted to other persons through blood

transfusion (Rios et al, 2006). From in vivo and in vitro studies, it can be postulated that

the increased incidence of fatal encephalitis in thioglycollate-treated susceptible HeJ

mice was due to the greater numbers of WNV-infected cells present in the peritoneum

of these mice. Consequently, this may induce higher and early viraemia as well as

higher serum TNFα production compared to control WNV-infected mice. The

increased TNFα production may have occurred around the same time as viraemia (2-3

days p.i), thus these two possibly act synergistically to cause a more severe disease

outcome.

The pathogenic role of macrophages following WNV infection in susceptible HeJ mice

was further supported by the adoptive transfer of infected thioglycollate-elicited

macrophages (Chapter 5). Similar to thioglycollate pretreatment, adoptive transfer of

WNV-infected macrophages also increased mortality and decreased ATD in HeJ mice.

Therefore, in human infections, although DCs are infected and responsible for

transporting virus to the lymph nodes following natural infection via mosquito bites,

macrophages may still have an important contribution to the dissemination of

flaviviruses and subsequent development of flavivirus-induced encephalitis. The

CHAPTER 8

GENERAL DISCUSSION

227

flavivirus-infected monocytes/macrophages then could migrate from the blood into the

peripheral tissues and the CNS, disseminating the virus throughout the body and

contributing to local tissue inflammation. Combined, in vivo and in vitro data clearly

indicate that macrophages play an important role in the pathogenesis of WNV in

susceptible mice during i.p. challenge. In contrast, their role in KUNV and MVEV

infection appears to be less significant.

Thioglycollate-elicited macrophages differ from resident peritoneal macrophages in

terms of types of cell population, activation state as well as morphology. While the

resident peritoneal macrophages are relatively homologous cell populations, mainly

consisting of mature macrophages, the thiogycollate-elicited macrophages are

heterogeneous cell populations which include immature, inflammatory macrophages

recruited from the circulating and marginal pools (van Furth, 1981). Thus, although it

was shown in this study that among thioglycollate-elicited macrophages, there are cells

that could support flavivirus infection, particularly WNV, it is not known what

particular macrophage sub-types could act as ‘Trojan Horses’ to disseminate the virus to

other organs. Previous work in this laboratory was conducted to look at the

permissiveness of different tissue macrophages to WNV infection (Pantelic, 2004).

While alveolar and bone marrow-derived macrophages have low susceptibility, resident

peritoneal macrophages can support WNV replication (Pantelic, 2004). Data from this

current study clearly suggested that in addition to the resident peritoneal macrophages,

thioglycollate treatment elicits additional cells that could play a role in distributing

WNV to other peripheral tissues and the brain.

One of the important findings of this study was that the resistance of DUB mice to i.p.

infection with WNV was not affected by any of the various treatments used in this

project to modulate the host non-specific immunity (macrophage), acquired immunity

(T cell) or the integrity of BBB. Treatments with thioglycollate (macrophage

enrichment in peritoneum), clodronate (macrophage depletion), SDS or LPS (breaching

of BBB), and depletion of CD4 and CD8+ T cells did not induce morbidity in resistant

DUB mice following i.p. WNV challenge most probably because the flavivirus

resistance gene is expressed in the peripheral tissues. This led to the unavailability of

highly permissive cells/tissues to support flavivirus replication and hence highlighting

the dominant role of Flvr gene over other host defense mechanisms in the protection of

i.p. flavivirus infection.

CHAPTER 8

GENERAL DISCUSSION

228

The second part of this project was focused on identifying underlying mechanisms that

could contribute to the outcome of infection during i.c. KUNV and MVEV in

susceptible HeJ and resistant DUB mice. Although KUNV was not neuroinvasive, i.c.

infection of this virus resulted in high incidence of fatal encephalitis of susceptible HeJ

and resistant DUB mice. HeJ and DUB mice succumbed to i.c. KUNV infection on day

5 and day 9 p.i., respectively. In contrast, i.c. MVEV inoculation only caused morbidity

to susceptible HeJ mice (died on day 6 p.i.) but not to resistant DUB mice. Although

incomplete protection in flavivirus resistant mice during certain i.c. flavivirus infections

have been reported earlier (Sabin, 1954; reviewed in Brinton and Perelygin, 2003,

Shueb, 2002), the cause of death of these mice has never been fully elucidated.

It was shown in this study that different factors were associated with the death of

susceptible and resistant mice during i.c. infection. This finding is significant since it

suggests that different pathogenic pathways are involved in different mouse strains

although similar fatal encephalitis could be observed. Death of susceptible HeJ mice

following KUNV and MVEV i.c. challenge was associated with high viral titres, severe

brain tissue inflammation and robust proinflammatory and Th1 cytokine production

(Chapter 6 and 7). Apoptosis of brain cells was found not to be associated with severe

KUNV and MVEV infection in HeJ as well as DUB mice as the occurrence of apoptosis

was not abundant.

Further flow cytometry analysis on four different types of brain mononuclear cells

indicated that CD11b+ cells were the predominant mononuclear cell population in the

brains of both susceptible HeJ and resistant DUB mice i.c. infected with either KUNV

or MVEV. Cells expressing CD11b+ surface markers may include resting microglia,

activated microglia as well as peripherally recruited macrophages and NK cells. The

next common cell type found was brain CD8+ T cells although the numbers varied

during different flavivirus infection. In both strains of mouse, KUNV induced more

infiltration of CD8+ T cells while MVEV caused more recruitment of CD4+ T cells, B

cells and microglia/macrophages (Chapter 7). The predominant accumulation of CD8+

T cells compared to CD4+ T cells in the brains of infected mice was in accordance with

studies on other flaviviruses (Liu et al, 1989a; Wang et al, 2003b). The accumulation of

CD8+ T cells in the CNS is intriguing, since lysis of CNS cells by cytotoxic T cells can

be fatal especially if this involves post mitotic cells such as neurons. It was found in this

CHAPTER 8

GENERAL DISCUSSION

229

project that in vivo KUNV and MVEV infection induced a greater increase in the

expression of MHC class I than MHC class II. This may explain the greater recruitment

of CD8+ T cells than CD4+ T cells to the brain.

Extensive studies have established that an intact immune system in general is required

for control of flavivirus infections (Chambers and Diamond, 2003). Mice with some

defects of the immune response become more vulnerable to virus infection.

Interestingly, in this study, CD4+ T cells had only a small contribution to KUNV and

MVEV pathogenesis in HeJ mice, as evidenced by T cell depletion studies. This result

is not in accordance with the findings recently reported on the requirement of CD4+ T

cells to sustain CD8+ T cell levels in the brain and in controlling WNV NY strain (Sitati

and Diamond, 2006), a virus that belongs to the same lineage as KUNV (WNV lineage

I). Similar crucial role of CD4+ T cells has been reported during i.c. infection of

neurotropic JHM strain of mouse hepatitis virus, whereby the absence of CD4+ T cells

induced apoptosis of CD8+ T cells in the CNS (Hickey, 1999; Stohlman et al, 1998).

CD4+ T cells could also contribute to the host antiviral defense by facilitating antibody

response, directing inflammatory/antiviral cytokine production and promoting memory

response (Mullbacher et al, 2003). The importance of CD4+ T cells in eliminating

infection and protecting the host from lethal disease has also been reported during

infection with measles virus, LCMV, rotavirus, Herpes simplex virus type 2 and

influenza virus, although the CD4+ T cells-dependent mechanisms vary significantly

between these viruses (Fehr et al, 1998; Jennings et al, 1991; Belz et al, 2002).

In contrast, CD8+T cells had no apparent role during i.c. KUNV infection in HeJ mice,

while they were neuroprotective in MVEV-infected HeJ. In other flavivirus infections,

CD8+ T cells also have disparate roles, further emphasizing that the role of T cells in

development of flavivirus-induced diseases varies, depending on mouse strain, route of

inoculation, virus strain, and dose of virus (Shrestha et al, 2004; Wang et al, 2003b;

reviewed in King et al, 2007). T cells may have both a protective and

immunopathological effect in mice infected with low doses of WNV Sarafend, as mice

lacking functional CD8+ T cells or depleted of these cells displayed increased mortality

but survived longer compared to control mice (Wang et al, 2003b). In contrast, T cells

were detrimental during MVEV and high doses of WNV Sarafend infection (Wang et

al, 2003b; Regner et al, 2001; Licon Luna et al, 2002). It remains to be investigated

what other inflammatory cells could possibly contribute to the fatal encephalitis of

CHAPTER 8

GENERAL DISCUSSION

230

KUNV and MVEV-infected susceptible HeJ mice since severe brain inflammation was

observed in these mice but with no pathogenic T cell role.

A strong brain Th1 cytokine response was elicited in HeJ mice infected with either

viruses, evidenced by the high levels of IFN and TNF. Nevertheless, the role of these

cytokines during KUNV and MVEV pathogenesis in HeJ mice is not known and would

be an interesting subject for future studies. In MVEV 1-51 i.v. infection, IFNαβ has

been demonstrated to be critical for protection against fatal encephalitis (Lobigs et al,

2003a). IFNαβ knockout mice had increased and persistent viraemia with subsequent

100% mortality observed after MVEV i.v. challenge. In contrast, IFNγ and NOS-2

knockout mice only displayed marginally increased susceptibility to the same virus

infection (Lobigs et al, 2003a).

The mechanism/event leading to earlier death of KUNV-infected HeJ mice than

MVEV-infected HeJ mice is yet to be elucidated. This is quite interesting given that

MVEV replicated to greater levels in brains of HeJ mice than KUNV. Furthermore, the

reasons for the incomplete KUNV clearance in sick DUB mice are also unknown. It is

possible that KUNV has different neurotropisms than MVEV in mice, infecting either

neurons in different part of the brain and/or in some inflammatory cells.

Hypochromatism, vacuolisation, and neuronophagia of neurons were evident in the

thalamus of KUNV-infected HeJ mice but not MVEV-infected HeJ mice, suggesting

that rigorous virus replication at this site may occur, leading to structural or functional

damage of the neurons. Involvement of macrophages/microglia has been reported in

several CNS-related diseases. For instance, immune-mediated demyelination induced by

mouse hepatitis virus strain JHM is associated with macrophage infiltration. Microglia

activation is also associated with a range of other diseases such as Alzheimers,

Parkinsons and HIV-associated dementia (Vilhardt, 2005). These activated

microglia/macrophages could have detrimental effects through the release of a myriad

of cytokines, free radicals and other mediators which could be toxic to the cells when

overproduced (Hanisch, 2002). Increased iNOS, IL-1β, IL-6, TNFα and MCP-1

expression have been implicated in microglia activation and neuronal death during JEV

infection in mice (Ghoshal et al, 2007). Recently, Wang and co-workers (2006) found

that some brain cells carrying cell surface marker CD11b+ were positive for WNV

when analysed by immunostaining. Thioglycollate-elicited macrophages of HeJ and

DUB mice supported in vitro KUNV replication better than MVEV, as previously

CHAPTER 8

GENERAL DISCUSSION

231

shown in Chapter 4. This suggests the possibility that macrophages and/or microglia to

act as reservoirs for KUNV in the brain. This may contribute to the severity of infection

in KUNV-infected DUB mice and to the earlier death of HeJ mice infected with KUNV

compared to MVEV. To further elucidate the role of microglia/macrophages during

KUNV and MVEV infection, depletion of blood borne macrophages using clodronate or

perivascular microglia/macrophages using mannosylated clodronate should be

performed (Bauer et al, 1995; Polfliet et al, 2001).

The Flv gene is responsible for limiting flavivirus infection and spread to other tissues

while host immune response usually involves in viral clearance. In this study, it was

revealed that the death of i.c. KUNV-infected DUB mice was not due to the ability of

KUNV to abrogate the expression and action of the flavivirus resistance gene, Flvr,

since a similar reduction of brain viral titres in resistant DUB mice following i.c.

infection with either KUNV or MVEV was observed. In fact, death of KUNV-infected

DUB mice was rather associated with an incomplete KUNV clearance. KUNV was

detected at low titres at the time of death, a similar finding to that reported during i.c.

French neurotropic YFV infection in RV mice (Sabin, 1954).

It was further showed in this project that a fatal infection in resistant DUB mice during

i.c. KUNV challenge could be attributed to an immunopathological disease, partly

caused by T cells. KUNV induced higher numbers of brain CD8+ T cells in resistant

DUB mice that persisted at the late stage of infection (day 7 to 9 p.i.) while brain CD8+

T cell levels declined at similar time points after i.c. MVEV challenge. CD8+ T cell

persistence has been reported during TMEV infection. SJL/J mice which are susceptible

to TMEV infection failed to clear the virus, leading to persistent infection in the brain

and consequently to the accumulation and retention of virus specific CD8+ T cells

(Lyman et al, 2004). From T cell depletion studies, CD8+ T cells were shown to have a

larger contribution to the development of fatal encephalitis in DUB mice infected with

KUNV than CD4+ T cells, possibly due to the higher numbers of the former cells in the

brain compared to the latter cells. Alternatively, CD4+ T cells might not be as important

as CD8+ T cells in the pathogenesis of KUNV. This finding bears a great significance

since this was the first time that the immunopathological role of T cells has been

described in resistant DUB mice. This finding also defines the cause of death of KUNV-

infected DUB mice. Interestingly, the results clearly showed that although KUNV

caused greater recruitment of brain CD8+ T cells in both susceptible HeJ and resistant

CHAPTER 8

GENERAL DISCUSSION

232

DUB mice, these cells have disparate role in different mouse strains. In sharp contrast,

T cells were neuroprotective during i.c. MVEV infection in resistant DUB mice since

mice lacking these cells exhibited signs of sickness. Depletion of a single subset of T

cells intriguingly did not render DUB mice susceptible to MVEV infection, suggesting

that CD4+ T cells and CD8+ T cells act in concert and perhaps able to compensate for

one another’s absence.

Although they can be pathogenic, T cells were shown to be necessary for viral clearance

following i.c. flavivirus infections in resistant DUB mice. This was supported by the

significantly higher brain titres in mice that lacked T cells compared to control mice.

This result is in accordance with findings reported by other investigators (reviewed in

Chambers and Diamond, 2003; Wang et al, 2004).

CD8+ T cells assume effector functions either by cytolysis or by production of IFN

(non-cytolytic mechanism) (Chesler and Reiss, 2002). CD8+ T cells-mediated cytolysis

is involved in limiting WNV Sarafend and WNV NY infection in mice (Wang et al,

2004b; Shrestha et al, 2006). However, given that apoptosis was not apparent during

KUNV infection in DUB mice, CD8+ T cells may not exert detrimental effect on DUB

mice via cytolysis of infected cells. In fact, based on the data attained in this study, T

cells probably inflict fatal encephalitis of KUNV-infected DUB mice through IFNγ

production. This was supported by the significantly higher brain IFNγ levels observed

in KUNV-infected DUB mice compared to MVEV (Student t test, p < 0.0003) at the

time when mice inoculated with the former virus displayed signs of sickness (day 7

p.i.). Further flow cytometric analysis confirmed that CD8+T cells were the major

producer of IFNγ at this time point. Future experimental work using IFNγ neutralising

antibodies or knockout mice are required to verify the immunopathogenic contribution

of IFNγ in KUNV-infected resistant DUB.

IFNγ is a pleiotropic cytokine and an important component of the cytokine-mediated

immune response to viral infections (Boehm et al, 1997). IFN may be involved in the

regulation of the host immune system, stimulation of antigen presentation MHC class I

and II, recruitment of leucocytes and control of cell proliferation and apoptosis (Boehm

et al, 1997). Contribution of this cytokine to immunopathological disease or host

recovery varies, depending on several aspects including genetic background of mouse

model, virus strain, route of inoculation and virus dose. Studies with several neurotropic

CHAPTER 8

GENERAL DISCUSSION

233

viruses indicated that IFNγ emerged more as an important host antiviral rather than a

pathogenic factor. Secretion of IFN by CD8+ T cells was shown to be essential for the

survival of hosts infected with poxvirus and Semliki Forest virus (Ramshaw et al, 1997;

Alsharifi et al, 2006). Furthermore, IFN was also involved in the clearance of LCMV,

mouse hepatitis virus, neuroadapted Sindbis virus and Borna virus (Bartholdy et al,

2000; Parra et al, 1999; Burdeinick-Kerr and Griffin, 2005; Hausmann et al, 2005). In

flavivirus infection, the role of IFN is paradoxical. The route of virus inoculation and

virus strain seems to affect the role of this cytokine in WNV infection. IFN does not

have any crucial role during i.v. infection of WNV Sarafend and KUNV since the lack

of IFN only marginally increased the susceptibility of mice (Wang et al, 2006)

However, IFN may be directly or indirectly associated with the immunopathological

disease of mice i.p. challenged with WNV Sarafend, as evidenced by the enhanced

survival rates (3 fold) of infected IFN knockout mice (King et al, 2003). In contrast,

increased vulnerability to virus infection was reported in mice lacking IFN production

or signalling following WNV NY strain inoculation by the s.c. route (Shrestha et al,

2006). Mortality increased to 90% in infected IFN knockout mice compared with only

30% death rate observed in wild type mice. Furthermore, the lack of IFN correlated

with higher levels of viraemia and viral replication in lymphoid tissues. This suggests

that during s.c. WNV NY infection, IFN has an early antiviral role that provides

protection in the peripheral tissues, thereby preventing virus invasion of the CNS

(Shrestha et al, 2006). However, none of these studies were performed in flavivirus

resistant mice or in susceptible mice of the C3H background. In fact, this is the first

study in which KUNV was shown to cause death to flavivirus resistant mice and CD8+

T cells that produced high levels of IFN contributed to the severity of the disease.

Interestingly, 7 days after i.c. KUNV and MVEV infection in DUB mice, although the

IFNγ outputs were remarkably different, the numbers of IFNγ producing-CD8+ T cells

were comparable (Table 7.11). This suggests that KUNV could induce higher IFNγ

production per CD8+ T cells than MVEV. Additionally, brain CD11b+ cells which

consisted largely of microglia/macrophages could contribute to the differential output of

IFNγ since CD11b+ cell population capable of producing IFNγ were more than double

in number in KUNV-infected than MVEV-infected DUB mice. However, on day 9 p.i.,

despite the higher CD8+ T cell numbers in the CNS of KUNV-infected, compared to

MVEV-infected DUB mice (more than double), similar brain IFNγ production was

CHAPTER 8

GENERAL DISCUSSION

234

observed during both infections. The reason for this is not readily explainable. It is

possible that as KUNV titres drastically declined from day 7 p.i. onwards, there were

less proinflammatory cytokines and infectious virus in the CNS. This may have led to

lower MHC expression on infected cells and APCs, as suggested by the lower

expression of these molecules seen on CD11b+ cells 9 days after infection (Section

7.2.1.3). Since activated T cells only perform their effector functions upon encountering

MHC-presented peptide on target cells (Boehm et al, 1997), the lacked of MHC

expression probably led to less interaction of T cells with target cells and eventually to

reduced IFNγ production. Additionally, two distinct signals delivered by APCs are

required for T cell activation, namely the peptide/MHC and co-stimulatory signals.

Interaction of CD28 on T cells with B7 on APCs generates one of the most important

co-stimulatory signals for T cell activation (reviewed in Boehm et al, 1997). However,

upon activation, T cells can also express CTLA-4 which is known to have high avidity

for B7 and binding of this ligand/receptor may send an inactivation signal to curtail T

cell responses. This may limit the effector role of activated T cells and consequently it

could influence the production of IFNγ in DUB mice 9 days after KUNV infection.

However, further studies are required to clarify this. During TMEV strain DA and strain

BeAN infection, the different level and avidity of virus-specific CD8+ T cells has been

suggested to contribute to the different outcome of disease in mice (Kang et al, 2002).

The activation status, level and avidity of virus-specific T cells during i.c. KUNV versus

MVEV infection however is unknown and would be an interesting subject for future

work.

In conclusion, data presented in this thesis have advanced our understanding on the

pathogenesis of three closely related flaviviruses; KUNV, WNV and MVEV in cell

culture and mouse models. As evidenced by the current study, virulence of flaviviruses

must be assessed independently or separately in different models of infection and cannot

be assumed to be similar even between very closely related flaviviruses. Each of these

viruses has different neuroinvasiveness and neurovirulence traits in susceptible versus

resistant mice. Macrophages were shown to have a dual role in i.p. WNV infection in

susceptible HeJ mice, both in host innate defense and as sites of WNV replication. In

addition to supporting flavivirus replication, macrophages may contribute to WNV

pathogenesis in susceptible mice by production of TNFα that is likely to alter the BBB

permeability. Although KUNV is a non-neuroinvasive virus, it is highly neurovirulent

in both susceptible and resistant mice. Thus, KUNV may pose a danger to humans if it

CHAPTER 8

GENERAL DISCUSSION

235

escapes the host antiviral immunity and enters the brain. This scenario may happen, for

example, in immuno-compromised people, although in this study, modulation of

macrophages (using thioglycollate and clodronate) and BBB permeability (using LPS

and SDS) in mice did not increase the virulence of KUNV in adult mice following i.p.

challenge.

Intracerebral infection with KUNV and MVEV induced different polarisation of the

host immune response in susceptible and resistant mice, which ultimately contributed to

different outcomes of infection, particularly in resistant mice. The results described in

this thesis provide further insight and important information regarding the mechanisms

of KUNV and MVEV pathogenesis in susceptible and resistant mice with a C3H

background. In susceptible mice, although a strong immune response was observed in

the brain during i.c. KUNV and MVEV infection, T cells did not have a significant

immunopathological role in development of fatal encephalitis. In contrast, T cells,

particularly CD8+ T cells, were shown to greatly contribute to the death of KUNV-

infected resistant DUB mice. The different numbers of recruited immune cells in the

brains of resistant mice could be partly attributed to the varying amounts of

proinflammatory cytokines such as IFN type I and TNFα produced following KUNV

and MVEV infection. CD8+ T cells exert a damaging effect on resistant DUB mice,

probably through excessive production of IFNγ. It remains to be answered why KUNV

cannot be cleared completely from the brains of infected resistant mice, what factors are

involved in regulating and polarising the recruited inflammatory cells and what immune

cells other than T cells contribute to the death or survival of KUNV and MVEV-

infected HeJ and DUB mice. The disparity in immune response and pathogenesis

observed when mice were challenged with different flaviviruses indicates that the

immune responses generated by the host-virus interactions cannot be generalized, even

between closely related flaviviruses (Wang et al, 2006).

REFERENCES

236

9.0 REFERENCES

Abbott, N.J., Ronnback, L.R.S., and Hanson, E. (2005). Astrocytes-endothelial

interactions at the blood-brain barrier. Nat. Rev. Neurosci. 7: 41-53.

Abraham, S., and Manjunath, R. (2006). Induction of classical and nonclassical MHC-

I on mouse brain astrocytes by Japanese encephalitis virus. Virus Research, 119: 216-

220.

Adam, D.O., and Hamilton, T.A. (1984). The cell biology of macrophage activation.

Ann. Rev. Immunol. 2: 283-318.

Aloise, F. (2001). Immune function of microglia. GLIA 36: 165-179.

Alsharifi, M., Lobigs, M., Simon, M.M., Kersten, A., Muller, K., Koskinen, A., Lee,

E., and Mullbacher, A. (2006). NK cell-mediated immunopathology during an acute

viral infection of the CNS. Eur. J. Immunol. 36: 887-896.

Alvarez, D.E., De Lella Ezcurra, A.L., Fucito, S., and Gamarnik, A.V. (2005). Role of

RNA structures present at the 3’UTR of dengue on translation, RNA synthesis, and

viral replication. Virology. 339: 200-212.

An, J., Zhou, D.S., Zhang, J.L., Morida, H., Wang, J.L., and Yasui, K. (2004).

Dengue-specific CD8+ T cells have both protective and pathogenic roles in dengue

virus infection. Immunol. Letters. 95: 167-174.

Anderson, J.R. (2001). The mechanisms of direct, virus-induced destruction of

neurons. Curr. Top.Microbiol. Immunol. 253: 15-33.

Anderson, R. (2003). Manipulation of cell surface macromolecules by flaviviruses.

Adv. Virus Res. 59: 229-274.

Andrews, D.M., Matthews, V.B., Sammels, L.M., Carrello, A.C., and McMinn, P.C.

(1999). The severity of Murray Valley encephalitis in mice is linked to neutrophil

infiltration and inducible nitric oxide synthase activity in the central nervous system.

J. Virol. 73: 8781-8790.

Asensio, V.C., and Campbell, I.L. (2001). Chemokines and viral diseases of the

REFERENCES

237

central nervous system. Adv. Virus Res. 56: 127-173.

Bachmann, M.F. and Kopf, M. (2002). Balancing protective immunity and

immunopathology. Curr. Opin. Immunol. 14: 413-419.

Bajetto, A., Bonavia, R., Barbero, S., and Schettini, G. (2002). Characterization of

chemokines and their receptors in the central nervous system: physiopathological

implications. J. Neurochem. 82: 1311-1329.

Barret, A.D.T., Cubitt, W.D., and Dimmock, N.J. (1984). Defective interfering

particles of Semliki forest virus are smaller than particles of standard virus. J. Gen.

Virol. 65: 2265-2268/

Bartholdy, C., Christensen, J.P., Wodarz, D., and Thomsen, A.R. (2000). Persistent

virus infection despite chronic cytotoxic T-lymphocyte activation in gamma

interferon-deficient mice infected with lymphocytic choriomeningitis virus. J. Virol.

74: 10304-10311.

Bauer, J., Huitinga, I., Zhao, W., Lassman, H., Hickey, W.F., and Dijkstra, C.D.

(1995). The role of macrophages, perivascular cells and microglial cells in the

pathogenesis of experimental autoimmune encephalomyelitis. GLIA. 15: 437-446.

Beasley, D.W., Li, L., Suderman, M.T., and Barrett, A.D. (2002). Mouse

neuroinvasive phenotype of West Nile virus strains varies depending upon virus

genotype. Virology. 296: 17-23.

Becher, B., Prat, A., and Antel, J.P. (2000). Brain-Immune Connection: Immuno-

regulatory properties of CNS-resident cells. GLIA. 29: 293-304.

Belardelli, F., Gessani, S., Proietti, E., Locardi, C., Borghi, P., Watanabe, Y., Kawade,

Y., and Gresser, I. (1987). Studies on the expression of spontaneous and induced

interferons in mouse peritoneal macrophages by means of monoclonal antibodies to

mouse interferons. J. Gen. Virol. 68: 2203-2212.

Belz, G. T., Wodarz, D., Diaz, G., Nowak, M. A. and Doherty, P. C. (2002).

Compromised influenza virus-specific CD8-T-cell memory in CD4-T-cell deficient

mice. J. Virol. 76:12388–12393.

REFERENCES

238

Ben-Nathan, D., Huitinga, I., Lustig, S., van Rooijen, N., and Kobiler, D. (1996). West

Nile virus neuroinvasion and encephalitis induced by macrophage depletion in mice.

Arch Virol. 141: 459-469.

Beutler, B. (2004). Inferences, questions and possibilities in Toll-like receptor

signalling. Nature. 430: 257-263.

Bhatt, P.N., and Jacoby, R.O. (1976). Genetic resistance to lethal flavivrus

encephalitis. II. Effect of immunosuppression. J. Infect. Dis. 134: 166-173.

Bhatt, P.N., Johnson, E.A., Smith, A.L., and Jacoby, R.O. (1981). Genetic resistance

to lethal flaviviral encephalitis. III. Replication of Banzi virus in vitro and in vivo in

tissues of congenic susceptible and resistant mice. Arch. Virol. 69: 273-286.

Biewenga, J., van der Ende, M.B., Krist, L.F., Borst, A., Ghufron, M., and van

Rooijen, N. (1995). Macrophage depletion in the rat after intraperitoneal

administration of liposome-encapsulated clodronate: depletion kinetics and accelerated

repopulation of peritoneal and omental macrophages by administration of Freund's

adjuvant. Cell Tissue Res. 280: 189-196.

Binder, G.K., and Griffin, D.E. (2001). Interferon--mediated site-specific clearance

of Alphavirus from CNS neurons. Science. 293: 303-306.

Binder, G.K., and Griffin, D.E. (2003). Immune-mediated clearance of virus from the

central nervous system. Microbes Infection. 5: 439-448.

Blackwell, J.L., and Brinton, M.A. (1997). Translation elongation factor-1 alpha

interacts with the 3' stem-loop region of West Nile virus genomic RNA. J. Virol. 71:

6433-6444.

Boehm, U., Klamp, T., Groot, M., and Howard, J.C. (1997). Cellular responses to

interferon-. Annu. Rev. Immunol. 15: 749-795.

Bogdan, C. (2001). Nitric oxide and the immune response. Nat. Immunol. 2: 907-916.

Bradl, M., and Flugel, A. (2002). The role of T cells in brain pathology. Curr. Topics

Microb. Immunol. 265: 141-162.

Brault, A.C., Langevin, S.A., Bowen, R.A., Panella, N.A., Biggerstaff, B.J., Miller,

REFERENCES

239

B.R., and Komar, N. (2004). Differential virulence of West Nile strains for American

crows. Emerg. Infect.Dis. 10: 2161-2168.

Brehm, M.A., Selin, L.K., and Welsh, R.M. (2004). CD8 T cell responses to viral

infections in sequence. Cell.Microbiol. 6: 411-421.

Brinton, M.A. (1981). Isolation of a replication-efficient mutant of West Nile virus

from a persistently infected genetically resistant mouse cell culture. J. Virol. 39: 413-

421.

Brinton, M.A. (1983). Analysis of extracellular West Nile virus particles produced by

cell cultures from genetically resistant and susceptible mice indicates enhanced

amplification of defective interfering particles by resistant cultures. J. Virol. 46: 860-

870.

Brinton, M.A. (1986). Replication of Flaviviruses, p.327-365. In: S. Schlesinger, M.J.

Schlesinger (eds), The Togaviridae and Flaviviridae. Plenum Press, New York.

Brinton, M.A. (1997). Host susceptibility to viral disease, p.303-328. In: N.

Nathanson, R. Ahmed, F. Gonzalez-Scarano, et al, (eds.), Viral pathogenesis.

Lippincott-Raven Publishers, Philadelphia.

Brinton, M.A. (2001). Host factors involved in West Nile virus replication. Ann. NY

Acad. Sci. 951: 207-219.

Brinton, M.A. (2002). The molecular biology of West Nile Virus: a new invader of the

western hemisphere. Annu. Rev. Microbiol. 56: 371-402.

Brinton, M.A., and Perelygin, A.A. (2003). Genetic resistance to flaviviruses. Adv.

Virus Res. 60: 43-85.

Brinton, M.A., Arnheiter, H., and Haller, O. (1982). Interferon independence of

genetically controlled resistance to flaviviruses. Infect. Immun. 36: 284-288.

Brinton, M.A., Kurane, I., Mathew, A. Zeng, L., Shi, P.Y., Rothman, A., and Ennis,

F.A. (1998). Immune mediated and inherited defeces against flaviviruses. Clin.

Diag.Virol. 10: 129-139.

Brinton-Darnell, M., and Koprowski, H. (1974). Genetically determined resistance to

REFERENCES

240

infection with group B arboviruses. II.Increased production of interfering particles in

cell cultures from resistant mice. J. Infect. Dis. 129: 248-256.

Brinton-Darnell, M., Koprowski, H., and Lagerspetz, K. (1974). Genetically

determined resistance to infection with group B arboviruses. I. Distribution of

resistance gene among various populations and characteristics of gene expression in

vivo. J. Infect. Dis. 129: 240-247.

Broom, A.K., Wallace, M.J., Mackenzie, J.S., Smith, D.W., Hall, R.A. (2000).

Immunisation with gamma globulin to murray valley encephalitis virus and with an

inactivated Japanese encephalitis virus vaccine as prophylaxis against australian

encephalitis: evaluation in a mouse model. J. Med. Virol. 61: 259-265.

Brosnan, C.F., Bornstein, M.B., and Bloom, B.R. (1981). The effects of macrophage

depletion on the clinical and pathologic expression of experimental allergic

encephalomyelitis. J. Immunol. 126:614-620.

Burdeinick-Kerr, R., and Griffin, D.E. (2005). Gamma interferon-dependent,

noncytolytic clearance of Sindbis virus infection from neurons in vitro. J Virol. 79:

5374-5385.

Burke, D.S., and Monath, T.P. (2001). Flaviviruses, p.1043-1125. In: D.M. Knipe,

P.M. Howley, D.E. Griffin, et al. (eds.), Fields Virology. 4th

edition. Lippincott

Williams and Wilkins, Philadelphia.

Cabarrocas, J., Bauer, J., Piaggio, E., Liblau, R., and Lassmann, H. (2003). Effective

and selective immune surveillance of the brain by MHC class I-restricted cytotoxic T

lymphocytes. Eur. J. Immunol. 33: 1174-82.

Calisher, C.H., and Gould, E.A. Taxonomy of the virus family Flaviviridae. (2003).

Adv. Virus. Res. 59: 1-19.

Calisher, C.H., Karabatsos, N., Dalrymple, J., Shope, R.E., Porterfield, J.S.,

Westaway, E.G., and Brand, W.E. (1989). Antigenic relationships between

flaviviruses as determined by cross-neutralisation tests with polyclonal antisera. J.

Gen Virol. 70: 37-43.

Calvert, A.E., Huang, C.Y.H., Kinney, R.M., and Roehrig, J.T. (2006). Non-structural

REFERENCES

241

proteins of dengue-2 virus offer limited protection to interferon-deficient mice after

dengue 2 virus challenge. J. Gen. Virol. 87: 339-346.

Camenga, D.L., Nathanson, N., and Cole, G.A. (1974). Cyclophosphamide-potentiated

West Nile viral encephalitis: relative influence of cellular and humoral factors. J.

Infect. Dis. 130: 634-641.

Cardosa, M.J., Gordon, S., Hirsch, S., Springer, T.A., and Poterfield, J.S. (1986).

Interaction of West Nile virus with primary murine macrophages: Role of cell

activation and receptors for antibody and complement. J. Virol. 57: 952-959.

Cardosa, M.J., Poterfield, J.S., and Gordon, S. (1983). Complement receptor mediates

enhances flavivirus replication in macrophages. J.Exp. Med. 158: 258-263.

Castle, E., Nowak, T., Leidner, U., Wengler, G., and Wengler, G. (1985). Sequence

analysis of the viral core and the membrane associated proteins VI and NV2 of the

flavivirus West Nile and of the genome sequence for these proteins. Virology. 145:

227-236.

Chambers, T.J., and Diamond, M.S. (2003). Pathogenesis of flavivirus encephalitis.

Avd. Virus Res. 60: 273-342.

Chambers, T.J., Hahn, C.S., Galler, R., and Rice, C.M. (1990a). Flavivirus genome

organization, expression, and replication. Ann. Rev. Microbiol. 44: 649-688.

Chambers, T.J., Halevy, M., Nestorowicz, A., Rice, C.M., and Lustig, S. (1998). West

Nile virus envelope proteins: nucleotide sequence analysis of strains differing in

mouse neuroinvasiveness. J. Gen. Virol. 79: 2375-2380.

Chambers, T.J., Weir, R.C., Grakoui, A., McCourt, D.W., Bazan, J.F., Fletterick, R.J.,

and Rice, C.M. (1990b). Evidence that the N-terminal domain of nonstructural protein

NS3 from yellow fever virus is a serine protease responsible for site-specific cleavages

in the viral polyprotein. Proc. Natl. Acad. Sci. USA. 87: 8898-8902.

Chaturvedi, U.C., Dhawan, R., Khanna, M., and Mathur, A. (1991). Breakdown of the

blood-brain barrier during dengue virus infection of mice. J. Gen. Virol. 72: 859-866.

Cheeran, M.C., Hu, S., Sheng, W.S., Rashid, A., Peterson, P.K., and Lokensgard, J.R.

(2005). Differential responses of human brain cells to West Nile virus infection. J.

REFERENCES

242

Neurovirol. 11: 512-524.

Chen, C.J., Kuo, M.D., Chien, L.J., Hsu, S.L., Wang, Y.M., and Lin, J.H. (1997).

RNA-protein interactions: Involvement of NS3, NS5 and 3’ noncoding region of

Japanese encephalitis virus genomic RNA. J. Virol. 71: 3466-3473.

Chen, C.J., Liao, S.L., Kuo, M.D., and Wang, Y.M. (2000). Astrocytic alteration

induced by Japanese encephalitis virus infection. Neuroreport. 11:1933-1937.

Chen, Y.C., and Wang, S.Y. (2002). Activation of terminally differentiated human

monocytes/macrophages by dengue virus: productive infection, hierarchical

production of innate cytokines and chemokines, and the synergistic effect of

lipopolysaccharide. J. Virol. 76: 9877-9887.

Cheng, Y., King, N.J., and Kesson, A.M. (2004). Major histocompatibility complex

class I (MHC-I) induction by West Nile virus: involvement of 2 signalling pathways in

MHC-I up-regulation. J. Infect. Dis. 189: 658-668.

Chesler, D.A., and Reiss, C.S. (2002). The role of IFN-gamma in immune responses

to viral infections of the central nervous system. Cytokine Growth Factor Rev. 13:

441-454.

Chu, J.J., Choo, B.G., Lee, J.W. amd Ng, M.L. (2003). Actin filaments participate in

west Nile (Sarafend virus maturation process. J. Med. Virol. 71: 463-472.

Chu, J.J.H., and Ng, M.L. (2002). Trafficking mechanism of west Nile (Sarafend)

virus structural proteins. J. Med. Virol. 67: 127-136.

Chu, P.W.G., and Westaway, E.G. (1985). Replication strategy of Kunjin virus:

Evidence for recycling role of replicative form RNA as template in semi-conservative

and asymmetric replication. Virology. 140: 68-79.

Chung, K.M., Nybakken, G.E., Thompson, B.S., Engle, M.J., Marri, A., Fremont,

D.H., and Diamond, M.S. (2006). Antibodies against West Nile Virus nonstructural

protein NS1 prevent lethal infection through Fc gamma receptor-dependent and -

independent mechanisms. J. Virol. 80: 1340-1351.

Ciavarra, R.P., Buhrer, K., Van Rooijen, N., and Tedeschi, B. (1997). T cell priming

against vesicular stomatitis virus analyzed in situ: red pulp macrophages, but neither

REFERENCES

243

marginal metallophilic nor marginal zone macrophages, are required for priming

CD4+ and CD8+ T cells. J Immunol. 158: 1749-1755.

Clark, D.C., Lobigs, M., Lee, E., Howard, M.J., Clark, K., Blitvich, B.J. and Hall,

R.A. (2007). In situ reactions of monoclonal antibodies with a viable mutant of

Murray Valley encephalitis virus reveal an absence of dimeric NS1 protein. J. Gen.

Virol. 88: 1175-1183

Cobbold, S.P., Jayasuriya, A., Rash, A., Prospero, T.D. and Waldmann, H. (1984).

Therapy with monoclonal antibodies by elimination of T cell subsets in vivo. Nature.

312: 584-550.

Cockfield, S.M., Ramassar, V., Noujaim, J., van der Meide, P.H., Halloran, P.F.

(1993). Regulation of IFN-gamma expression in vivo. IFN-gamma up-regulates

expression of its mRNA in normal and lipopolysaccharide-stimulated mice. J.

Immunol. 150: 717-725.

Coia, G., Parker, M.D., Speight, G., Byrne, M.E., and Westaway, E.G. (1988).

Nucleotide and complete amino sequences of Kunjin virus: definitive gene order and

characteristics of the virus-specified proteins. J. Gen. Virol. 69: 1-21.

Cole, G.A., and Nathanson, N. (1968). Potentiation of experimental arbovirus

encephalitis by immunosuppressive doses of cyclophosphamine. Nature. 220: 399-

401.

Courageot, M.P., Catteau, A., and Despres, P. (2003). Mechanisms of Dengue virus-

induced cell death. Adv. Virus Res. 60: 157-186.

Dalgarno, L., Trent, D.W., Strauss, J.H., and Rice, C.M. (1986). Partial nucleotide

sequence of the Murray Valley encephalitis virus genome. Comparisons of the

encoded polypeptides with Yellow fever virus structural and non structural proteins. J.

Mol. Biol. 187: 309: 323.

Darnay, B.G., and Aggarwal, B.B. (1999). Signal transduction by tumor necrosis

factor and tumor necrosis factor related ligands and their receptors. Ann. Rheum. Dis.

58: I2-I13.

Darnell, M.B., Koprowski, H., and Lagerspetz, K. (1974). Genetically determined

REFERENCES

244

resistance to infection with group B arboviruses. I. Distribution of the resistance gene

among various mouse populations and characteristics of gene expression in vivo. J.

Infect. Dis. 129: 240-247.

Desai, A., Murali-Krishna, K., Ramireddy, K., Ravi, V., and Manjunath, R. (1997). In

vivo clearance of Japanese encephalitis virus by adoptively transferred virus specific

cytotoxic T lymphocytes. J. Biosci. 22: 33-45.

Desai, A., Shankar, S.K., Ravi, V., Chandramuki, A., and Gourie-Devi, M. (1995).

Japanese encephalitis virus antigen in the human brain and its topographic

distribution. Acta Neuropathol (Berl). 89: 368-373.

Despres, P., Flamond, M., Ceccaldi, P.-E., and Deubel, V. (1996). Human isolates of

Dengue type 1 virus induce apoptosis in mouse neuroblastoma cells. J. Virol. 70:

4090-4096.

Despres, P., Frenkiel, M.-P., Ceccaldi, P.-E., Duarte Dos Santos, C., and Deubel, V.

(1998). Apoptosis in the mouse central nervous system in response to infection with

mouse neurovirulent dengue viruses.J. Virol. 72: 823-829.

Deubel, V., Fiette, L., Gounon, P., Drouet, MT., Khun, H., Huerre, M., Banet, C.,

Malkinson, M., and Despres, P. (2001). Variations in biological features of West Nile

viruses. Ann. N. Y. Acad. Sci. 951: 195-206.

Di Marzio, P., Gessani, S., Locardi, C., Borghi, P., Baglioni, C., and Belardelli, F.

(1990). Effects of different biological response modifiers on interferon expression in

bacterial lipopolysaccharide (LPS)-responsive and LPS-hyporesponsive mouse

peritoneal macrophages. J. Gen. Virol. 71: 2585-2591.

Diamond, M.S. (2003). Evasion of innate and adaptive immunity by flavivirues.

Immunol. Cell Biol. 81: 196-206.

Diamond, M.S., and Harris, E. (2001). Interferon inhibits Dengue virus infection by

preventing translation of viral RNA through a PKR-independent mechanism.

Virology. 289: 297-311.

Diamond, M.S., Roberts, T.G., Edgil, D., Lu, B., Ernst, J., Harris, E. (2000).

Modulation of Dengue virus infection in human cells by alpha, beta and gamma

REFERENCES

245

interferons. J. Virol. 74: 4957-4966.

Diamond, M.S., Shrestha, B., Marri, A., Mahan, D., and Engle, M. (2003b). B cells

and antibody play critical roles in the immediate defense of disseminated infection by

West Nile encephalitis virus. J. Virol. 77: 2578-2586.

Diamond, M.S., Shrestha, B., Mehlhop, E., Sitati, E., and Engle, M. (2003a). Innate

and adaptive immune responses determine protection against disseminated infection

by West Nile encephalitis virus. Viral Immunol. 16: 259-278.

Dietzschold, B., Kao, M., Zheng, Y.M., Chen, Z.Y., Maul, G., Fu, Z.F., Rupprecht,

C.E., and Koprowski, H. (1992). Delineation of putative mechanisms involved in

antibody-mediated clearance of rabies virus from the central nervous system. Proc.

Natl. Acad. Sci. USA. 89: 7252-6. Erratum in: Proc. Natl. Acad. Sci. USA 89: 9365.

Doherty, R.L., Carley, J.G., Mackerras, M.J., and Marks, E.N. (1963). Studies of

arthropodborne virus infections in Queensland. III. Isolation and characterization of

virus strains from wild-caught mosquitoes in North Queensland. Aust. J Exp. Biol.

Med. Sci. 41: 17-39.

Dorries, R. (2001). The role of T-cell-mediated mechanisms in virus infections of the

nervous system. Curr. Top. Microbiol. Immunol. 253: 219-45.

Duane, J.G., and Roehrig, J.T. (2000). Arboviruses (Togaviridae and Flaviviridae),

p.579-600. In: L. Collier, A. Balows, M. Susswan et al., (eds.), Topley and Wilson’s

Microbiology and microbial infections 9th

edition, Oxford University Press, New

York.

Dulbecco, R. (1990). Interference with viral multiplication, p. 381-418. In: B.D.

Davis, R. Dulbecco, H.N., Eisen, and H.S. Ginsberg (eds.), Microbiology, Harper

International edition.

Elkon KB, Liu CC, Gall JG, Trevejo J, Marino MW, Abrahamsen KA, Song X, Zhou

JL, Old LJ, Crystal RG, Falck-Pedersen E. (1997). Tumor necrosis factor alpha plays a

central role in immune-mediated clearance of adenoviral vectors. Proc Natl Acad Sci

U S A. ;94 : 9814-9819.

Engle, M.J., and Diamond, M.S. (2003). Antibody prophylaxis and therapy against

REFERENCES

246

West Nile virus infection in wild type and immunodeficient mice. J. Virol. 77: 12941-

12949.

Fehr, T., Naim, H. Y., Bachmann, M. F., Ochsenbein, A. F., Spielhofer, P., Bucher,

E., Hengartner, H., Billeter, M. A., and Zinkernagel, R. M. (1998). T-cell independent

IgM and enduring protective IgG antibodies induced by chimeric measles viruses. Nat.

Med. 4: 945–948

Fink, J., Gu, F., and Vasudevan, S.G. (2006). Role of T cells, cytokines and antibody

in dengue fever and dengue haemorrhagic fever. Rev. Medic. Virol. 16: 263-275.

Fischer, H.G., and Reichmann, G. (2001). Brain dendritic cells and

macrophages/microglia in central nervous system inflammation. J. Immunol. 166:

2717-2726.

Fisher, H.G., Bonifas, U., and Reichmann, G. (2000). Phenotype and functions of

brain dendritic cells emerging during chronic infection of mice with toxoplasma

gondii. J. Immunol. 164: 4826-4834.

Fuhlbrigge, R.C., Chaplin, D.D., Kiely, J.M., and Unanue, E.R. (1987). Regulation of

interleukin 1 gene expression by adherence and lipopolysaccharide. J. Immunol. 138:

3799-802.

Garcia-Tapia, D., Loiacono, C.M., and Kleiboeker, S.B. (2006). Replication of West

Nile virus in equine peripheral blood mononuclear cells. Veterinary Immunol.

Immunopathol. 110: 229-244.

Gehrmann, J. (1996). Microglia: a sensor to threats in the nervous system? Res. Virol.

147: 79-88.

Ghoshal, A., Sulagna, D., Ghosh, S., Mishra M.K., Sharma, V., Koli, P., Sen, E., and

Basu, A. (2007). Proinflammatory mediators released by activated microglia induce

neuronal death in Japanese encephalitis. GLIA. 55: 483-496.

Glass, W.G., Lim, J.K., Cholera, R., Pletnev, A.G., Gao, J.L., and Murphy, P.M.

(2005). Chemokine receptor CCR5 promotes leucocyte trafficking to the brain and

survival in West Nile infection. J. Exp. Med. 202:1087-1098.

Gollins, S.W., and Porterfield, J.S. (1984). Flavivirus infection enhancement in

REFERENCES

247

macrophages: Radioactive and biological studies on the effect of antibody on viral

fate. J. Gen. Virol. 65: 1261-1272.

Gollins, S.W., and Porterfield, J.S. (1985). Flavivirus infection enhancement in

macrophages: an electron microscopic study of viral cellular entry. J. Gen. Virol.

66:1969-1982.

Goodman, G.T., and Koprowski, H. (1962a). Study of the mechanism of innate

resistance to virus infection. J. Cell. Comp. Physiol. 59: 333-373.

Goodman, G.T., and Koprowski, H. (1962b). Macrophages as a cellular expression of

inherited natural resistance. PNAS. 148:160-5.

Gould, L.H., and Fikrig, E. (2004). West Nile virus: a growing concern? J. Clin.

Invest. 113: 1102-1107.

Gouwy, M., Struyf, S., Proost, P., and Van Damme, J. (2005). Synergy in cytokine

and chemokine networks amplifies the inflammatory response. Cytokine and Growth

Factors Reviews. 16: 561-580.

Green, M.C. (1989). Catalogue of mutant genes and polymorphic loci, p. 8-274. In:

Green, M.C. (ed), Genetic Variants and Strains of the Laboratory mouse. Gustav

Fischer Verlag, Stuttgart, New York.

Gregg, A.R., Lee, C.G., Herman, G.E., and O’Brian, W.E. (1995). Endothelial nitric

oxide synthase (Nos3) maps to the proximal region of mouse chromosome 5. Mamm.

Genome. 6: 152.

Griffin, D.E. (1995). Arbovirses and the central nervous system. Springer Semin.

Immunopathol. 17: 121-132.

Griffin, D.E. (2003). Immune responses to RNA-virus infections of the CNS. Nature

Reviews Immunol. 3: 493-502.

Groschel, D., and Koprowski, H. (1965). Development of a virus-resistant inbred

mouse strain for the study of innate resistance to Arbo B viruses. Arch. Ges.

Virusforsch. 17:379-391.

Gubler, D.J. (2002). The global emergence/resurgence of arboviral disease as public

REFERENCES

248

health problem. Arch. Med. Res. 33: 330-342.

Guidotti, L.G., and Chisari, F.V. (2001). Noncytolitic control of viral infections by the

innate and adaptive immune response. Annu. Rev. Immunol. 19: 65-91.

Guo, J.T., Hayasi, J., and Seeger, C. (2005). West Nile virus inhibits the signal

transduction pathway of alpha interferon. J. Virol. 79: 1343-1350.

Haahr, S. (1971). The influence of poly I:C on the course of infection in mice

inoculated with West Nile virus. Arch. Gesamte Virusforch. 35: 1-9.

Halevy, M., Akov, Y., Ben-Nathan, D., Kobiler, D., Lachmi, B., and Lustig, S. (1994).

Loss of active neuroinvasiveness in attenuated strains of West Nile virus:

pathogenecity in immunocompetent and SCID mice. Arch. Virol. 137: 355-370.

Hall, R.A., Brand, T.N.H., Lobigs, M., Sangster, M.Y., Howard, M.J., and Mackenzie,

J.S. (1996). Protective immune responses to the E and NS1 proteins of Murray Valley

encephalitis virus in hybrids of flavivivirus-resistant mice. J. Gen. Virol. 77: 1287-

1294.

Hall, R.A., Broom, A.K., and Smith, D.W. (2002). The ecology and epidemiology of

Kunjin virus. Curr. Topic Microbiol. Immunol. 267: 253-270.

Hall, R.A., Khromykh, A.A., Mackenzie J.M., Scherret, J.H., Khromykh, T.I.

Mackenzie, J.S. (1999). Loss of dimerisation of the Nonstructural Protein NS1 of

Kunjin virus delays viral replication and reduces virulence in mice, but still allows

secretion of NS1. Virology. 264: 66-75.

Hall, R.A., Scherret, J.H., and Mackenzie, J.S. (2001). Kunjin Virus An Australian

variant of West Nile? Ann. NY Acad. Sci. 951: 153-160.

Haller, O., Arnheiter, H., Gresser, I., and Lindenmann, J. (1979). Genetically

determined interferon-independent resistance to influenza virus in mice. J. Exp. Med.

149: 601-612.

Haller, O., Arnheiter, H., Lindenmann, J., and Gresser, I. (1980). Host gene influences

sensitivity to interferon action selectively for influenza virus. Nature 283: 660-662.

Haller, O., Kochs, G., and Weber, F. (2006). The interferon response circuit: Induction

REFERENCES

249

and suppression by pathogenic viruses. Virology. 344: 119-130.

Halstead, S.B., and O'Rourke, E.J. (1977). Antibody-enhanced dengue virus infection

in primate leukocytes. Nature. 265: 739-741.

Hanisch, U.K. (2002). Microglia as a source and target of cytokines. Glia. 40: 140-

155.

Hanson, B., Koprowski, H., Baron, S., and Buckler, C.E. (1969). Interferon-mediated

natural resistance of mice to ArboB virus infection. Microbios. 1B:51-68.

Hase, T., Dubios, D.R., and Summers, P.L. (1990b). Comparative study of mouse

brains infected with Japanese encephalitis virus by intracerebral or intraperitoneal

inoculation. Int. J. Exp. Path. 71: 857-869.

Hase, T., Summers, P.L., and Dubous, D.R. (1990a). Ultrastructural changes of mouse

brains neurons infected with Japanese encephalitis virus. J. Exp. Pathol. 71: 493-505.

Hase, T., Summers, P.L., Eckels, K.H., and Baze, W.B. (1987). An electron and

immunoelectron microscopic study of dengue-2 virus infection of cultured mosquito

cells: maturation events. Arch. Virol. 92: 273-291.

Haskill, S., Johnson, C., Eierman, D., Becker, S., and Warren, K. (1988). Adherence

induces selective mRNA expression of monocyte mediators and proto-oncogenes. J.

Immunol. 140: 1690-1694.

Hausmann, J., Pagenstecher, A., Baur, K., Richter, K., Rziha, H.J., and Staeheli, P.

(2005). CD8 T cells require gamma interferon to clear borna disease virus from the

brain and prevent immune system-mediated neuronal damage. J. Virol. 79: 13509-

13518.

Hay, S., and Kannoroukis, G. (2002). A time to kill: viral manipulation of the cell

death program. J. Gen. Virol. 83: 1547-1564.

Heinz, F.X. (1986). Epitope mapping of flavivirus glycoproteins. Adv. Virus Res. 31:

103-168.

Heinz, F.X. (2003). Molecular aspects of TBE virus research. Vaccine. 21: S1/3-

S1/10.

REFERENCES

250

Heinz, F.X., and Allison, S.L. (2002). The machinery for flavivirus fusion with host

cell membranes. Curr. Opinion Microbiol. 4: 450-455.

Heinz, F.X., Collett, M.S., Purcell, R.H., et al. (2000). Family Flaviviridae, p.859-878.

In: M.H.V. van Reggenmortel, C.M. Fauquet, D.H.L. Bishop et al., (eds.), Virus

Taxonomy. Classification and Nomenclature of Viruses. 7th

report of the International

Committee for the Taxonomy of Viruses, Academic Press, San Diego, CA.

Hendriks, J.J., Teunissen, C.E., de Vries, H.E., and Dijkstra, C.D. (2005).

Macrophages and neurodegeneration. Brain Res. Brain Res. Rev. 48: 185-195.

Hickey, W.F. (1999). Leucocyte traffic in the central nervous system: the participants

and their roles. Semin. Immunol. 11: 125-137.

Higashi, Y., Sokowa, Y., Watanabe, Y., Kawade, Y., Ohno, S., Takaoka, C., and

Taniguchi, T. (1983). Structure and expression of a cloned cDNA for mouse

interferon-beta. J. Biochem. Chem. 258: 9522-9529.

Hill, A.B., Mullbacher, A., Parrish, C., Coia, G., Westaway, E.G., and Blanden, R.V.

(1992). Broad cross-reactivity with marked fine specificity in the cytotoxic T cell

response to flaviviruses. J. Gen. Virol. 73: 1115-1123.

Hober, D., Shen, L., Benyoucef, S., De Groote, D., Deubel, V., and Wattre, P. (1996).

Enhanced TNF production by monocytic-like cells exposed to dengue virus antigens.

Immunol. Letters. 53: 115-120.

Hooper, D.C., Sauder, C., Scott, G.S., Dietzschold, B., and Richt, J.A. (2002).

Immunopathology and immunoprotection in CNS virus infections: Mechanisms of

virus clearance from the CNS. Curr. Topics Microb. Immunol. 265: 163-182.

Hurrelbrink, R.J., and McMinn, P.C. (2001). Attenuation of Murray Valley

encephalitis virus by site-directed mutagenesis of the hinge and putative receptor-

binding regions of the envelope protein. J. Virol. 75: 7692-7702.

Hurrelbrink, R.J., and McMinn, P.C. (2003). Molecular determinants of virulence: The

structural and functional basis for flavivirus attenuation. Adv. Virus. Res. 60: 1-42.

Irani, D.N., and Griffin, D.E. (1996). Regulation of lymphocyte homing into the brain

REFERENCES

251

during viral encephalitis at various stages of infection. J. Immunol. 156: 3850-3857.

Irani, D.N., Lin, K.I., and Griffin, D.E. (1997). Regulation of brain-derived T cells

during acute central nervous system inflammation. J. Immunol. 158: 2318-2326.

Ishak, R., Tovey, D.G., and Howard, C.R. (1988). Morphogenesis of yellow fever

virus 17D in infected cell cultures. J. Gen. Virol. 169: 325-35.

Itano, A.A., and Jenkins, M.K. (2003). Antigen presentation to naïve CD4 T cells in

the lymph node. Nat. Immunol. 4: 733-739.

Jacoby, R.O., and Bhatt, P.N. (1976). Genetic resistance to lethal flavivirus

encephalitis I. Infection of congenic mice with Banzi virus. J. Infect. Dis. 134: 158-

165.

Jacoby, R.O., Bhatt, P.N., and Schwartz, A. (1980). Protection of mice from lethal

flavivirus encephalitis by adoptive transfer of splenic cells from donors infected with

Banzi virus. J. Infect. Dis. 141: 617-624.

Jennings, S. R., Bonneau, R. H., Smith, P. M., Wolcott, R. M. and Chervenak, R.

(1991). CD4-positive T lymphocytes are required for the generation of the primary but

not the secondary CD8-positive cytolytic T lymphocyte response to herpes simplex

virus in C57BL/6 mice. Cell. Immunol. 133:234–252.

Johnson, L.J., Halliday, G.M., and King, N.J.C. (2000). Langerhans cells migrate to

local lymph nodes following cutaneous infection with arbovirus. J. Invest. Dermatol.

114: 560-568.

Johnson, R.T., Burke, D.S., Elwell, M., Leake, C.J., Nisalak, A., Hoke, C.H., and

Lorsomrudee, W. (1985). Japanese encephalitis: immunocytochemical studies of viral

antigen and inflammatory cells in fatal cases. Ann. Neurol. 18: 567-573.

Kajaste-Rudnitski, A., Mashimo, T., Frenkiel, M.P., Guenet, J.L., Lucas, M., and

Despres, P. (2006). The 2’, 5’-oligoadenylate synthetase 1b is a potent inhibitor of

West Nile replication inside infected cells. J. Biol. Chem. 281: 4624-4637.

Kesson, A.M., and King, N.J.C. (2001). Transcriptional regulation of major

histocompatibility complex class I by flavivirus West Nile is dependent on NF-kB

REFERENCES

252

activation. J. Infect. Dis. 184: 947-954.

Kesson, A.M., Blanden, R.V., and Mullbacher, A. (1987). The primary in vivo

cytotoxic T cell response to the flavivirus, West Nile. J. Gen. Virol. 68: 2001-2006.

Kesson, A.M., Blanden, R.V., and Mullbacher, A. (1988). The secondary in vitro

murine cytotoxic T cell response to the flavivirus, West Nile. Immunol. Cell Biol. 66:

23-32.

Kesson, A.M., Cheng, Y., and King, N.J.C. (2002). Regulation of immune recognition

molecules by Flavivirus, West Nile. Viral. Immunol. 15: 273-283.

Khromykh, A.A., Meka, H., Guyatt, K.J., and Westaway, E.G. (2001). Essential role

of cyclization sequences in flavivirus RNA replication. J. Virol. 75: 6719-6728.

Kimura, T., and Griffin, D.E. (2000). The role of CD8+ T cells and major

histocompatibility complex class I expression in the central nervous system of mice

infected with neurovirulent Sindbis virus. J.Virol. 74: 6117-6125.

Kimura, T., and Griffin, D.E. (2003). Extensive immune-mediated hippocampal

damage in mice surviving infection with neuroadapted Sindbis virus. Virology. 311:

28-39.

King, N.J.C., Getts, D.R., Getts, M.T., Rana, S., Shrestha, B, and Kesson, A.M.

(2007). Immunopathology of flavivirus infections. Immunol. Cell Biol. 85: 33-42.

King, N.J.C., and Kesson, A.M. (1988). Interferon-independent increases in class I

major histocompatibility complex antigen expression follow flavivirus infection. J.

Gen. Virol. 69: 2535-2543.

King, N.J.C., and Kesson, A.M. (2003). Interaction of flaviviruses with cells of the

vertebrate host and decoy of the immune response. Immunol. Cell Biol. 81: 207-216.

King, N.J.C., Shrestha, N., and Kesson, A.M. (2004). Immuno modulation by

flaviviruses. Adv. Virus. Res. 60: 121-151.

Klein, R.S., Lin, E., Luster, A.D., Tollett, J., Samuel, M.A., Engle, M., and Diamond,

M.S. (2005). Neuronal CXCL10 directs CD8+ T-cell recruitment and control of West

Nile virus encephalitis. J. Virol. 79: 11457-11466.

REFERENCES

253

Kobiler, D., Lustig, S., Gozes, Y., Ben-Nathan, D., and Akov, Y. (1989). Sodium

dodecylsulphate induces a breach in the blood-brain barrier and enables a West Nile

virus variant to penetrate into mouse brain. Brain Res. 496: 314-316.

Kodukula, P., Liu, T., Rooijen, N.v., Jager, M.J., and Hendricks, R.L. (1999).

Macrophage control of herpes simplex virus type I replication in the peripheral

nervous system. J. Immunol. 162: 2895-2905.

Konishi, E., Yamaoka, M., Khin-Sane-Win, Kurane, I., Takada, K., Mason, P.W.

(1999). The anamnestic neutralizing antibody response is critical for protection of

mice from challenge following vaccination with a plasmid encoding the Japanese

encephalitis virus premembrane and envelope genes. J. Virol. 73: 5527-5534.

Kontny, U., Kurane, I., and Ennis, A. (1998). Gamma interferon augments Fc

receptor-mediated Dengue virus infection of human monocytic cells. J. Virol. 62:

3928-3933.

Kreil, T.R., and Eibl, M.M. (1995). Viral infection of macrophage profoundly alters

requirements for induction of nitric oxide synthesis. Virology. 212: 174-178.

Kreil, T.R., and Eibl, M.M. (1996). Nitric oxide and viral infection: NO antiviral

activity against a Flavivirus in vitro, and evidence for contribution to pathogenesis in

experimental infection in vivo. Virology 219: 304-306.

Kreil, T.R., Byrger, I., Bachmann, M., Fraiss, S., and Eibl, M.M. (1997). Antibodies

protect mice against challenge with tick-borne encephalitis virus (TBEV)-infected

macrophages. Clin. Exp. Immunol. 110: 358-361.

Kreutzberg, G.W. (1996). Microglia: a sensor for pathological events in the CNS.

Trends Neurosci. 19: 312-318.

Kunzi, M.S., and Pitha, P.M. (2000). The role of cytokines in viral infections, p.193-

210. In: L. Collier, A. Balows, M. Susswan, et al., (eds.), Topley and Wilson’s

Microbiology and microbial infections 9th

edition, Oxford University Press, New

York.

Kurane, I. (2002). Immune responses to Japanese encephalitis virus. Curr. Topic.

Immunol. Microbiol. 267: 91-104.

REFERENCES

254

Kurane, I., Hebblewaite, D., Brandt, W.E., and Ennis, F.A. (1984). Lysis of dengue

virus-infected cells by natural cell-mediated cytotoxicity and antibody-dependent cell-

mediated cytotoxicity. J. Virol. 52: 223-230.

Kurane, I., Innis, B.L., Nimmannitya, S., Nisalak, A.M., Meager, A., Janus, J., and

Ennis, F.A. (1991). Activation of T lymphocytes in dengue virus infections: high

levels of soluble interlukin 2 receptor, soluble CD4, soluble CD8, interleukin 2, and

interferon-γ in sera of children with dengue. J Clin. Invest. 88: 1473-1480.

Labrada, L., Liang, X.H., Zheng, W., Johnston, C., and Levine, B. (2002). Age-

dependent resistance to lethal alphavirus encephalitis in mice: analysis of gene

expression in the central nervous system and identification of a novel interferon-

inducible protective gene, mouse ISG12. J. Virol. 76: 11688-11703.

Lanciotti, R.S., Ebel, G.D., Deubel, V., Kerst, A.J., Murri, S., Meyer, R., Bowen, M.,

McKinney, N., Morrill, W.E., Crabtree, M.B., Kramer, L.D., and Roehrig, J.T. (2002).

Complete genome sequence and phylogenetic analysis of West Nile virus strains

isolated from the United States, Europe and the Middle East. Virology. 298: 96:105.

Lanciotti, R.S., Roehrig, J.T., Deubel, V., Smith, J., Parker, M., Steele, K., Crise, B.,

Volpe, K.E., Crabtree, M.B., Scherret, J.H., Hall, R.A., MacKenzie, J.S., Cropp, C.B.,

Panigrahy, B., Ostlund, E., Schmitt, B., Malkinson, M., Banet, C., Weissman, J.,

Komar, N., Savage, H.M., Stone, W., McNamara, T., and Gubler, D.J. (1999). Origin

of the West Nile virus responsible for an outbreak of encephalitis in the northeastern

United States. Science. 286: 2333-2337.

Lathbury, L.J., Allan J.E., Shellam, G.R. and Scalzo, A.A. (1996). Effect of host

genotype in determining the relative roles of natural killer cells and T cels in

mediating protection against murine cytomegalovirus infection. J. Gen. Virol. 77:

2603-2613.

Lee, C.G.L., Gregg, A.R., and O’Brien, W.E. (1995). Localisation of the neuronal

form of nitric oxide synthetase to mouse chromosome 5. Mamm. Genome. 6: 56-57.

Lee, E., Fernon, C., Simpson, R., Weir R.C., Rice, C.M. and Dalgarno, L. (1990).

Sequence of the 3’ half of Murray Valley encephalitis virus and mapping of the non-

structural NS1, NS3 and NS5. Virus Genes. 4: 197-213.

REFERENCES

255

Lee, E., and Lobigs, M. (2002). Mechanism of virulence attenuation of

glycosaminoglycan-binding variants of Japanese encephalitis virus and Murray Valley

encephalitis virus. J. Virol. 76: 4901-4911.

Lee, J.W.M., Chu, J.J.H., and NG, M.L. (2006). Quantifying the specific binding

between West Nile vurs envelope domain III protein and the cellular receptor αVβ3

integrin. J Biol. Chem. 281: 1352-1360.

Levine, B. (2002). Apoptosis in viral infections of neurons: a protective or

pathological host response? Curr. Topic Microb. Immunol. 265: 95-118.

Li, J., Bhuvanakantham, K., Howe, J., and Ng, M.L. (2005). Identifying the region

influencing the cis-mode of maturation of West Nile (Sarafend) virus using chimeric

infectious clones. Biochem. Biophys. Res. Commun. 334: 714-720.

Li, Y.M., Baviello, G., Vlassara, H., and Mitsuhashi, T. (1977). Glycation products in

aged thioglycollate medium enhance the elicitation of peritoneal macrophages. J.

Immunol. 201: 183-188.

Li, Y.M., Baviello, G., Vlassara, H., and Mitsuhashi, T. (1977). Glycation products in

aged thioglycollate medium enhance the elicitation of peritoneal macrophages. J.

Immunil. Meth. 201: 183-188.

Liao, C.L., Lin, Y.L., Wang, J.J. Huang, Y.L., Yeh, C.T., Ma, S.H., and Chen, L.K.

(1997). Effect of enforced expression of human bcl-2 on Japanese encephalitis virus-

induced apoptosis in cultured cells. J. Virol. 71: 5963-5971.

Libraty, D.H., Pichyangkul, S., Ajariyakhajorn, C., Endy, T.P., and Ennis, F.A.

(2001). Human dendritic cells are activated by dengue virus infection: Enhancement

by gamma interferon and implications for disease pathogenesis. J. Virol. 75: 3501-

3508.

Licinio, J., Prolo, P., McCann, S.M., and Wong, M.L. (1999). Brain iNOS: current

understanding and clinical implications. Mol. Med. Today 5: 225-232.

Licon Luna, R.M., Lee, E., Mullbacher, A., Blanden, R.V., Langman, R., and Lobigs,

M. (2002). Lack of both Fas ligand and perforin protects from flavivirus-mediated

encephalitis in mice. J. Virol. 76: 3202-3211.

REFERENCES

256

Liebert, U.G. (2001). Slow and persistent virus infections of neurones – a compromise

for neuronal survival. Curr. Top. Microbiol. Immunol. 253: 35-60.

Liehne, P.F., Anderson, S., Stanley, N.F., Liehne, C.G., Wright, A.E., Chan, K.H.,

Leivers, S., Britten, D.K., and Hamilton, N.P. (1976). Isolation of Murray Valley

encephalitis virus and other arboviruses in the Ord River Valley 1972-1976. Aust. J.

Exp. Biol. Med. Sci. 59: 347-356.

Ligon, B.L. (2005). Dengue feverand dengue haemorrhagic fever: A review in history,

transmission, treatment and prevention. Semin. Pediatr. Infect. Dis. 16: 60-65.

Lin, Y.L., Huang, Y.L., Ma, S.H., Yeh, C.T., Chiou, S.Y., Chen, L.K., and Liao, C.L.

(1997). Inhibition of Japanese encephalitis virus infection by nitric oxide: antiviral

effect of nitric oxide on RNA virus replication. J. Virol. 71: 5227-5235.

Lindenbach, B.D., and Rice, C.M. (1999). Genetic interaction of flavivirus

nonstructural proteins NS1 and NS4A as a determinant of replicase function. J.Virol.

736: 4611-4621.

Lindenbach, B.D., and Rice, C.M. (2001). Flaviviridae: The viruses and their

replication, p.991-1041. In: D.M. Knipe, P.M. Howley, D.E. Griffin et al. (eds.),

Fields Virology. 4th

edition, LippinCort Williams and Wilkins, Philadelphia.

Lindenbach, B.D., and Rice, C.M. (2003). Molecular biology of flaviviruses.

Adv. Virus Res. 59: 23-61.

Liou, M.L., and Hsu, C.Y. (1998). Japanese encephalitis virus is transported across the

cerebral blood vessels by endocytosis in mouse brain. Cell Tissue Res. 293: 389-394.

Liu, T., and Chambers, T.J. (2001). Yellow Fever virus encephalitis: Properties of the

brain-associated T-cell response during virus clearance in normal and gamma

interferon-deficient mice and requirement for CD4+ lymphocytes. J. Virol. 75: 2107-

2118.

Liu, W.J., Chen, H.B., and Khromykh, A.A. (2003). Molecular and functional analysis

of Kunjin virus infectious cDNA clones demonstrate the essential roles for NS2A in

virus assembly and for a nonconservative residue in NS3 in RNA replication. J. Virol.

77: 7804-7813.

REFERENCES

257

Liu, W.J., Wang, X.J., Clark, D.C., Lobigs, M., Hall, R.A., and Khromykh, A.A.

(2006). A single amino acid substitution in the West Nile virus nonstructural protein

NS2A disables its ability to inhibit alpha/beta interferon induction and attenuates virus

virulence in mice. J. Virol. 80: 2396-2404.

Liu, Y., Blanden, R.V., and Mullbacher, A. (1989a). Identification of cytolytic

lymphocytes in West Nile virus infected murine central nervous system. J. Gen. Virol.

70: 565-573.

Liu, Y., King, N.J.C., Kesson, A.M., Blanden, R.V., and Mullbacher, A. (1989b).

Flavivirus infection up-regulates the expression of class I and class II major

histocompatibility antigens on and enhances T cell recognition of astrocytes in vitro. J.

Neuroimmunol. 21: 157-168.

Lo, M.K., Tilgner, M., Bernard, K.A., and Shi, P.Y. (2003). Functional analysis of

mosquito-borne flavivirus conserved sequence elements within 3’ untranslated region

of West Nile virus by use of a reporting replicon that differentiates between viral

translation and RNA replication. J. Virol. 77: 10004-10014.

Lobigs M., Mullbacher A., and Regner M. (2003b). MHC class I up-regulation by

flaviviruses: Immune interaction with unknown advantage to host or pathogen.

Immunol. Cell. Biol. 81: 217-223.

Lobigs, M., and Lee, E. (2004). Inefficient signalase cleavage promotes efficient

nucleocapsid incorporation into budding flavivirus membranes. J. Virol. 78: 178-186.

Lobigs, M., Arthur, C.E., Mullbacher, A., and Blanden, R.V. (1994). The flavivirus

nonstructural protein NS3 is a dominant source of cytotoxic T cell peptide

determinants. Virology. 202: 195-201.

Lobigs, M., Mullbacher, A., Wang, Y., Pavy, M., and Eva, L. (2003a). Role of type I

and type II interferon responses in recovery from infection with an encephalitic

flavivirus. J.Gen. Virol. 84: 567-572.

Lobigs, M., Usha, R., Nestorowicz, A., Marshall, I.D., Weir, R.C., and Dalgarno, L.

(1990). Host cell selection of Murray Valley encephalitis virus variants altered at an

RGD sequence in the envelope protein and in mouse virulence. Virology. 176: 587-

REFERENCES

258

595.

Loke, H., Bethell, D.B., Phuong, C.X., Dung, M., Schneider, J., White, N.J., Day,

N.P., Farrar, J., and Hill, A.V. (2001). Strong HLA class

I-restricted

T

cell

responses

in

dengue hemorrhagic fever:

a

double-edged

sword?

J. Infect. Dis. 184: 1369-1373.

Lorenz, I.C., Allison, S.L., Heinz, F.X., and Helenius, A. (2002). Folding and

dimerization of tick-borne encephalitis virus envelope proteins prM and E in the

endoplasmic reticulum. J. Virol. 76: 5480-5491.

Lorenz, I.C., Kartenbeck, J., Mezzacasa, A., Allison, S.L. Heinz, F.X., and Helenius,

A. (2003). Intracellular assembly and secretion of recombinant subviral particles from

tick-borne encephalitis virus. J. Virol. 77: 4370-4382.

Lucas, M., Mashimo, T., Frenkiel, M.P., Simon-Chazottes, D., Montagutelli, X.,

Ceccaldi, P.E., Guenet, J.L., and Despres, P. (2003). Infection of mouse neurones by

West Nile virus is modulated by interferon-inducible 2’-5’ oligoadenylate synthetase

1b protein. Immunol. Cell Biol. 81: 230-236.

Lucas, S.M., Rothwell, N.J., and Gibson, R.M. (2006). The role of inflammation in

CNS injury and disease. British J. Pharmacology. 147: S232-S240.

Lucey, D.R., Clerici, M., and Shearer, G.M. (1996). Type 1 and Type 2 Cytokine

Dysregulation in Human Infectious, Neoplastic, and Inflammatory Diseases. Clin.

Microb. Rev. 9: 532-562.

Lustig, S., Danenberg, H.D., Kafri, Y., Kobiler, D., and Ben-Nathan, D. (1992). Viral

neuroinvasion and encephalitis induced by lipopolysaccharide and its mediators. J.

Exp. Med. 176: 707-712.

Lyman, M.A., Myoung, J., Mohindru, M., and Kim, B.S. (2004). Qualitative, not

quantitative, differences in CD8+ T cell responses to Theiler’s murine

encephalomyelitis virus between resistant C57BL/6 and susceptible SJL/J mice. Eur.

J. Immunol. 34: 2730-2739.

Lynch, C.J., and Hughes, T.P. (1936). The inheritance of susceptibility to yellow fever

encephalitis in mice. Genetics. 21: 104-112.

Mackenzie, J.M., and Westaway, E.G. (2001). Assembly and maturation of the

REFERENCES

259

flavivirus Kunjin appear to occur in the rough endoplasmic reticulum and along the

secretory pathway, respectively. J. Virol. 75: 10787-10799.

Mackenzie, J.M., Jones, M.K., and Young, P.R. (1996). Immunolocalization of the

Dengue virus nonstructural glycoprotein NS1 suggests role in viral RNA replication.

Virology. 220: 232-240.

Mackenzie, J.S., Barret, A.D.T., and Deubel, V. (2002a). The Japanese encephalitis

serological group of Flaviviruses: a brief introduction to the group. Curr. Top.

Microbiol. Immunol. 267: 1-10.

Mackenzie, J.S., Gubler, D.J., and Peterson, L.R. (2004). Emerging flaviviruses: the

spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat.

Med. Supp. 10: 98-109.

Mackenzie, J.S., Johansen, C.A., Ritchie, S.A., van den Hurk, A.F., and Hall, R.A.

(2002b). Japanese encephalitid as an emerging virus: the emergence and spread of

Japanese encephalitis virus in Australasia. Curr. Top. Microbiol. Immunol. 267: 49-74.

Mandl, C.W., Guirakhoo, F., Holzmann, H., Heinz, F.X., and Kunz, C. (1989).

Antigenic structure of the flavivirus envelope protein E at the molecular level, using

tick-borne encephalitis virus as a model. J. Virol. 63: 564-71.

Mangada, M.M., Endy, T.P., Nisalak, A., Chunsuttiwat, S., Vaughn, D.W., Libraty,

D.H., Green, S., Ennis, F.A., and Rothman, A.L. (2002). Dengue-specific T cell

responses in peripheral blood mononuclear cells obtained prior to secondary dengue

virus infections in Thai schoolchildren. J. Infect. Dis. 185: 1697-1703.

Mashimo, T., Lucas, M., Simon-Chazottes, D., Frenkiel, M.P., Montagutelli, X.,

Ceccaldi, P.E., Deubel, V., Guenet, J.L., Despres, P.A. (2002). Nonsense mutation in

the gene encoding 2'-5'-oligoadenylate synthetase/L1 isoform is associated with West

Nile virus susceptibility in laboratory mice.

Proc. Natl. Acad. Sci. USA. 99:11311-6.

Mathur, A., Arora, K.L., and Chaturvedi, U.C. (1983). Host defence mechanisms

against Japanese Encephalitis virus infection in mice. J. Gen Virol. 64: 805-811.

Mathur, A., Khanna, N., and Chaturvedi, U.C. (1992). Breakdown of blood-brain

REFERENCES

260

barrier by virus-induced cytokine during Japanese encephalitis virus infection. Int. J.

Exp. Path. 73: 603-611.

McMahon, E.J., Bailey, S.L., and Miller, S.D. (2006). CNS dendritic cells: critical

participants in CNS inflammation? Neurochem. Int. 49: 195-203.

McMinn, P.C. (1997). The molecular basis of virulence of the encephalitogenic

flaviviruses. J. Gen. Virol. 78: 2711-2722.

McMinn, P.C., Dalgarno, L., and Weir, R.C. (1996). A comparison of the spread of

Murray Valley encephalitis viruses of high or low neuroinvasiveness in the tissues of

swiss mice after peripheral inoculatin. Virology. 220: 414-423.

McMinn, P.C., Lee, E., Hartley, S., Roehrig, J.T., Dalgarno, L., and Weir, R.C.

(1995). Murray Valley encephalitis virus envelope protein antigenic variants with

altered hemagglutination properties and reduced neuroinvasiveness in mice. Virology.

211: 10-20.

Mims, C.A. 1960. Intracerebral injections and the growth of viruses in the mouse

brain. Br. J. Exp. Pathology. 41: 52-59.

Mizuno, T., Goto, Y., Bab, K., Masuda, K., Ohno, K., and Tsujimoto, H. (2001).

TNF-induced cell death in Feline Immunodeficiency virus-infected cells is mediated

by the caspase cascade. Virology. 287: 446-455.

Momburg, F., Mullbacher, A., and Lobigs, M. (2001). Modulation of transporter

associated with antigen processing (TAP)-mediated peptide import into the

endoplasmic reticulum by Flavivirus infection. J. Virol. 75: 5663-5671.

Monath, T.P., and Borden, E.C. (1971). Effects of thorotrast on humoral antibody,

viral multiplication, and interferon during infection with St. Louis encephalitis virus in

mice. J. Infect. Dis. 123: 297-300.

Mongkolsapaya, J., Duangchinda, T., Dejnirattisai, W., Vasanawathana, S., Avirutnan,

P., Jairungsri, A., Khemnu, N., Tangthawornchaikul, N., Chotiyarnwong, P., Sae-Jang,

K., Koch, M., Jones, Y., McMichael, A., Xu, X., Malasit, P., and Screaton, G. (2006).

T cell responses in dengue hemorrhagic fever: are cross-reactive T cells suboptimal? J.

Immunol. 176: 3821-3829.

REFERENCES

261

Mullbacher, A., and Lobigs, M. (1995). Up-regulation of MHC class I by flavivirus-

induced peptide translocation into the endoplasmic reticulum. Immunity. 3: 207-214.

Mullbacher, A., Lobigs, M., and Lee, E. (2003). Immunology of mosquito-borne

encephalitic flavivirus. Adv. Virus. Res. 60: 87-120.

Mullbacher, A., Lobigs, M., Hia, R.T., Tran, T., Stehle, T., and Simon, M.M. (2002).

Antigen-dependent release of IFN-γ by cytotoxic T cells up-regulates Fas on target

cells and facilitates exocytosis-independent specific target cell lysis. J. Immunol. 169:

145-150.

Munoz-Jordan, J.L., Sanchez-Burgos, G.G., Laurent-Rolle, M., and Garcia-Sastre, A.

(2003). Inhibition of interferon signaling by dengue virus. PNAS. 100: 14333-14338.

Murali-Krishna, K., Ravi, V., and Manjunath, R. (1994). Cytotoxic T lymphocytes

raised against Japanese encephalitis virus: Effector cell phenotype, target specificity

and in vitro virus clearance. J. Gen. Virol. 75: 799-807.

Murali-Krishna, K., Ravi, V., and Manjunath, R. (1996). Protection of adult but not

newborne mice against lethal intracerebral challenge with Japanese encephalitis virus

by adoptively transferred virus-specific cytotoxic T lymphocytes: requirement for

L3T4+ T cells. J. Gen. Virol. 77: 705-714.

Murray, J.M., Aaskov, J.G., and Wright, P.J. (1993). Processing of the dengue virus

type 2 proteins prM and C-prM. J. Gen. Virol. 74: 175-182.

Muylaert, I.R., Chambers, T.J., Galler, R.G., and Rice, C.M. (1996). Mutagenesis of

the N-linked glycosylation sites of yellow fecer virus NS1 protein: Effects on virus

replication and mouse neurovirulence. Virology. 222: 159-168.

Nash, A.A., and Usherwood, E.J. (2000). The immune response to viral infections,

p.173-191. In: L. Collier, A. Balows, M. Susswan, et al., (eds.), Topley and Wilson’s

Microbiology and microbial infections 9th

edition, Oxford University Press, New

York.

Nathan, C. (1992). Nitric oxide as a secretory product of mammalian cells. FASEB 6:

3051-3064.

Nathanson, N., and Cole, G.A. (1970). Fatal Japanese encephalitis virus infection in

REFERENCES

262

immunosuppressed spider monkeys. Clin. Exp. Immunol. 6: 161-166.

Navarro-Sanchez, E., Depres, P., and Cedillo-Barron, L. (2005). Innate immune

responses to Dengue virus. Arch. Medic.Res. 36: 425-435.

Neves-Souza, P.C., Azeredo, E.L., Zagne, S.M., Valls-de-Souza, R., Reis, S.R.,

Cerqueira, D.I., Nogueira, R.M., and Kubelka, C.F. (2005). Inducible nitric oxide

synthase (iNOS) expression in monocytes during acute Dengue Fever in patients and

during in vitro infection. BMC Infect Dis. 5: 64.

Ng, M.L., and Chu, J.H. (2002). Interaction of West Nile and Kunjin viruses with

cellular components during morphogenesis. Curr. Topic. Microbiol. Immunol. 267:

353-372.

Ng, M.L., and Lau, L.C. (1988). Possible involvement of receptors in the entry of

Kunjin virus into Vero cells. Arch. Virol. 100: 199-211.

Ng, M.L., Sreenivasan, V., and Mulders, J.J.L. (1994). Flavivirus West Nile

(Sarafend) egress at the plasma membrane. Arch. Virol. 137: 303-313.

Ng, M.L., Tan, S.H., and Chu, J.J.H. (2001). Transport and budding at two distinct

sites of visible nucleocapsids of West Nile (Sarafend) virus. J. Med. Virol. 65: 758-

764.

O'Garra, A. (1998). Cytokine induce the development of functionally heterogenous T

helper subsets. Immunity. 8: 275-283.

Pantelic, L. (2004). Peritoneal macrophages as an in vitro model of flavivirus

replication and their role in the pathogenesis of WN virus. Ph.D. thesis. Discipline of

Microbiology, School of Biomedical and Chemical Sciences, The University of

Western Australia.

Pantelic, L., Sivakumaran, H., and Urosevic, N. (2005). Differential induction of

antiviral effects against West Nile virus in primary mouse macrophages derived from

flavivirus-susceptible and congenic resistant mice by alpha/beta interferon and poly(I-

C). J. Virol. 79: 1753-1764.

Parquet, M.C., Kumatori, A., Haseba, F., Mathenge, E.G., and Igarashi, A. (2002). St.

Louis encephalitis virus induced pathology in cultured cells. Arch. Virol. 147: 1105-

REFERENCES

263

1119.

Parra, B., Hinton, D.R., Marten, N.W., Bergmann, C.C., Lin, M.T., Yang, C.S., and

Stohlman SA. (1999). IFN-gamma is required for viral clearance from central nervous

system oligodendroglia. J. Immunol. 162: 1641-7.

Perelygin, A.A., Scherbik, S.V., Zhulin, I.B., Stockman, B.M., Li, Y., and Brinton,

M.A. (2002). Positional cloning of the murine flavivirus resistance gene. Proc. Natl.

Acad. Sci. USA. 99: 9322-9327.

Perelygin, A.A., Scherbik, S.V., Zhulin, I.B., Stockman, B.M., Li, Y., and Brinton,

M.A. (2002). Positional cloning of the murine flavivirus resistance gene. Proc. Natl.

Acad. Sci. USA. 99: 9322-9327.

Perez, A.B., Garcia, G., Sierra, B., Alvarez, M., Vazquez, S., Cabrera, M.V.,

Rodriquez, R., Rosario, D., Martinez, E., Denny, T., and Guzman, M.G. (2004). IL-10

levels in Dengue patients: Some findings from the exceptional epidemiological

conditions in Cuba. J. Med. Virol. 73: 230-234.

Perry, V.H., and Gordon, S. (1988). Macrophage and microglia in the nervous system.

TINS. 11: 273-277.

Pletnev, A.G., Bray, M., Huggins, J., Lai, C.J. (1992). Construction and

characterization of chimeric tick-borne encephalitis/dengue type 4 viruses.

Proc. Natl. Acad. Sci. USA. 89: 10532-10536.

Polfliet, M.M., Goede, P.H., van Kesteren-Hendrikx, E.M., van Rooijen, N., Dijkstra,

C.D., and van den Berg, T.K. (2001). A method for the selective depletion of

perivascular and meningeal macrophages in the central nervous system. J.

Neuroimmunol. 116: 188-195.

Pope, M., Marsden, P.A., Cole, E., Sloan, S., Fung, L.S., Ning, Q., Ding, J.W.,

Leibowitz, J.L., Phillips, M.J., and Levy, G.A. (1998). Resistance to murine hepatitis

virus strain 3 is dependent on production of nitric oxide.

J. Virol. 72: 7084-70s90.

Prikhod’ko, G.G., Prikhod’ko, E.A., Pletnev, A.G., and Cohen, J.I. (2002). Langat

flavivirus protease NS3 binds caspase 8 and induces apoptosis. J.Virol. 76: 5701-5710.

REFERENCES

264

Raghupathy, R., Chaturverdi, U.C., Al-Sayer, H., Elbishbishi, E.A., Agarwal, R.,

Nagar, R., Kapoor, S., Misra, A., Mathur, A., Nusrat, H., Azizieh, F., Khan, M.A., and

Mustafa, A.S. (1998). Elevated levels of IL-8 in dengue hemorrhagic fever. J. Med.

Virol. 56: 280-285.

Raivich, G., and Banati, R. (2004). Brain microglia and blood-derived macrophages:

molecular profiles and functional roles in multiple sclerosis and animal models of

autoimmune demyelinating disease. Brain Res. Brain Res. Rev. 46: 261-281.

Ramshaw, I.A., Ramsay, A.J., Karupiah, G., Rolph, M.S., Mahalingam, S., and Ruby,

J.C. (1997). Cytokines and immunity to viral infections. Immunol Rev. 159: 119- 35.

Ravi, V., Parida, S., Desai, A., Chandramuki, A., Gourie-Devi, M., and Grau, G.E.

(1997). Correlation of tumor necrosis factor levels in the serum and cerebrospinal fluid

with clinical outcome in Japanese encephalitis patients. J. Med. Virol. 51: 132-136.

Redwine, J.M., and Evans, C.F. (2002). Markers of central nervous system glia and

neurons in vivo during normal and pathological conditions. Curr. Top Microbiol.

Immunol. 265: 119-140.

Reed, L.J., and Muench, H. (1938). A simple method of estimating fifty per cent

endpoint. Am. J. Hyg. 27: 493-497.

Regner, M., Lobigs, M., Blanden, R.V., and Mullbacher, A. (2001). Effector cytolytic

function but not IFNγ production in cytotoxic T cells triggered by virus-infected target

cells in vitro. Scan. J. Immunol. 54: 366-374.

Reiss, C.S., and Komatsu, T. (1998). Does nitric oxide play a critical role in viral

infections. J. Virol. 72: 4547-4551.

Reiss, C.S., Chesler, D.A., and Hodges, J. (2002). Innate immune responses in viral

encephalitis. Curr. Topics Microb. Immunol. 265: 63-94.

Rice, C.M. (1996). Flaviviridae: the viruses and their replication, p.931-960. In:

Fields, B.N., Knipe, D.M., and Howley, P.M. (eds.), Fields Virology, 3rd

edition.

Lippincott-Raven Publishers, Philadelphia.

Rice, C.M., Lenches, E.M., Eddy, S.R., Shin, S.J., Sheets, R.L., and Strauss, J.H.

(1985). Nucleotide sequence of yellow fever virus: implications for flavivirus gene

REFERENCES

265

expression and evolution. Science. 229: 726-33.

Rios, M., Zhang, M.J., Grinev, A., Srinivasan, K., Daniel, S., Wood, O., Hewlett, I.K.,

and Dayton, A.I. (2006). Monocytes-macrophages are potential target in human

infection with West Nile virus through blood transfusion. Transfusion. 46: 659-667.

Roehrig, J.T., Hunt, A.R., Johnson, A.J., and Hawkes, R.A. (1989). Synthetic peptides

derived from the deduced amino acid sequence of the E-glycoprotein of Murray

Valley encephalitis virus elicit antiviral antibody.Virology. 171: 49-60.

Rossi, C.P., Delcroix, M., Huitinga, I., McAllister, A., van Rooijen, N., Claassen, E.,

and Brahic, M. (1997). Role of macrophages during Theiler's virus infection. J. Virol.

71: 3336-3340.

Sabin, A.B. (1952a). Genetic, horomonal and age factors in natural resistance to

certain viruses. N Y Acad. Sci. 54: 936-945.

Sabin, A.B. (1952b). Nature of inherited resistance to viruses affecting the nervous

system. Proc. Natl. Acad. Sci. USA. 38: 540-546.

Sabin, A.B. (1954). Genetic factors affecting susceptibility and resistance to viruse

diseases of the nervous system. Res. Publ. Assoc. Res. Nerv. Ment. Dis. 33: 57-66.

Saija, A., Princi, P., Trombetta, D., Lanza, M., and De Pasquale, A. (1997). Changes

in the permeability of the blood-brain barrier following sodium dodecyl sulphate

administration in the rat. Exp. Brain. Res. 115: 546-551.

Sambhi, S.K., Kohonen-Corish, M.R.J., and Ramshaw, I.A. (1991). Local production

of tumor necrosis factor encoded by recombinant vaccinia virus is effective in

controlling viral replication in vivo. Proc. Natl. Acad. Sci. USA. 88: 4025-4029.

Samuel, C.E. (2001). Antiviral action of interferons. Clin. Microbiol. Rev. 14: 778-

809.

Samuel, C.E. (2002). Host genetic variability and West Nile virus susceptibility. Proc.

Natl. Acad. Sci. USA. 99: 11555-11557.

Samuel, M.A., and Diamond, M.S. (2005). Alpha/beta interferon protects against

lethal West Nile virus infection by restricting cellular tropism and enhancing neuronal

REFERENCES

266

survival. J. Virol. 79: 13350-13361.

Sangster, M.Y., and Shellam, G.R. (1986). Genetically controlled resistance to

flaviviruses within the house mouse complex of species. Current Top. Microbiol.

Immunol. 127: 313-318.

Sangster, M.Y., Heliams, D.B., MacKenzie, J.S., and Shellam, G.R. (1993). Genetic

studies of flavivirus resistance in inbred strains derived from wild mice: Evidence for

a new resistance allele at the flavivirus resistance locus (Flv). J. Virol. 67: 340-347.

Sangster, M.Y., Mackenzie, J.S., and Shellam, G.R. (1998). Genetically determined

resistance to flavivirus infection in wild Mus musculus domesticus and other

taxonomic groups in the genus Mus. Arch. Virol. 143: 697-715.

Sangster, M.Y., Urosevic, N., Mansfield, J.P., MacKenzie, J.S., and Shellam, G.R.

(1994). Mapping the Flv locus controlling resistance to flaviviruses on mouse

Chromosome 5. J. Virol. 68: 448-452.

Santana, M.A., and Rosenstein, Y. (2003). What it takes to become an effector T cells:

The process, the cells involved, and the mechanisms. J Cell. Physiol. 195: 392-401.

Satoh, J., Lee, Y.B., and Kim, S.U. (1995). T-cell costimulatory molecules B7-1

(CD80) and B7-2 (CD86) are expressed in human microglia but not in astrocytes in

culture. Brain Res. 704: 92-96.

Saxena, S.K., Singh, A., and Mathur, A. (2000). Antiviral effect of nitric oxide during

Japanese encephalitis virus infrction. Int. J. Exp. Path. 81: 165-172.

Scalzo, A.A., Fitzgerald, N.A., Simmons, A., La Vista, A.B., and Shellam, G.R.

(1990). Cmv-1, a genetic locus that controls murine cytomegalovirus replication in the

spleen. J. Exp. Med. 171: 1469-1483.

Scalzo, A.A., Lyons, P.A., Fitzgerald, N.A., Forbes, C.A., Yokoyama, W.M., and

Shellam, G.R. (1995). Genetic mapping of Cmv1 in the region of mouse chromosome

6 encoding the NK gene complex-associated loci Ly49 and musNKR-P1. Genomics.

27: 435-441.

Scherbik, S.V., Paranjape, J.M., Stockman, B.M., Silverman, R.H., and Brinton, M.A.

(2006). RNase L plays a role in the antiviral response to West Nile virus. J. Virol. 80:

REFERENCES

267

2987-2999.

Scherret, J.H., Poidinger, M., Mackenzie, J.S., Broom, A.K., Deubel, V., Lipkin, W.I.,

Briese, T., Gould, E.A., and Hall, R.A. (2001). The relationships between West Nile

and Kunjin viruses. Emerg. Infect. Dis. 7: 697-705.

Schneider-Schaules, S., Dunster, L.M., and ter Meulen, V. (2000). Infections of the

central nervous system. In: Collier L, Balows A, Susswan M, et al., editors. Topley

and Wilson’s Microbiology and microbial infections 9th

edition. New York: Oxford

University Press; 2000:p835-871.

Shaw, D.R., and Griffin Jr, F.M. (1982). Thioglycollate-elicited mouse peritoneal

macrophages are less efficient then resident macrophages in antibody-dependent cell-

mediated cytolysis. J. Immunol. 128: 433-440.

Shaw, G.D., Boll, W., Taira, H., Mantei, N., Lengyel, P., and Weissmann, C. (1983).

Structure and expression of cloned murine IFN-alpha genes. Nucleic Acids Res. 11:

555-573.

Shawar, S.M., Vyas, J.M., Rodgers, P.R., and Rich, R.R. (1994). Antigen presentation

by major histocompatibility complex class I-B molecules. Annu. Rev. Immunol. 12:

839-880.

Shellam, G.R., Sangster, M.Y., and Urosevic, N. (1998). Genetic control of host

resistance to flavivirus infection in animals. Rev. Sci. Tech. Off. Int. Epiz. 17: 231-248.

Shen, J., Devery, J.M., and King, N.J.C. (1995a). Adherence status regulates the

primary cellular activation responses to the flavivirus West Nile. Immunology. 84:

245-264.

Shen, J., Devery, J.M., and King, N.J.C. (1995b). Early induction of interferon-

dependent virus-specific ICAM-1 (CD54) expression by flavivirus in quiescent but not

proliferating fibroblast. Virology. 208: 437-449.

Shen, J., T-To, S.S., Schrieber, L., and King, N.J.C. (1997). Early E-selectin, VCAM-

1, ICAM-1 and late major histocompatibility complex antigen induction on human

endothelial cells by flavivirus and comodulation of adhesion molecule expression by

immune cytokines. J. Virol. 9323-9332.

REFERENCES

268

Shi, P.Y., Tilgner, M., and Lo, M.K. (2002). Construction and characterization of

subgenomic replicons of New York strain of West Nile virus. Virology, 296: 219-233.

Shieh, W.J., Guarner, J., Layton, M., Fine, A., Miller, J., Nash, D., Campbell, G.L.,

roehrig, J.T., Gubler, D.J., and Zaki S.R. (2000). The role of pathology in an

investigation of an outbreak of West Nile encephalitis in New York 1999. Emerg.

Infect. Dis. 6: 370-372.

Shirato, K., Kimura, T., Mizutani, T., Kariwa, H., and Takashima, I. (2004b).

Different chemokine expression in lethal and non-lethal murine West Nile virus

infection. J. Med. Virol. 74: 507-513.

Shirato, K., Miyoshi, H., Goto, A., Ako, Y., Ueki, T., Kariwa, H., and Takashima, I.

(2004a). Viral envelope protein glycosylation is a molecular determinant of the

neuroinvasiveness of the New York strain of West Nile virus. J. Gen. Virol. 85: 3637-

3645.

Shirato, K., Miyoshi, H., Kariwa, H., and Takashima, I. (2006). The kinetics of

proinflammatory cytokines in murine peritoneal macrophages infected with envelope

protein-glycosylated or non-glycosylated West Nile virus. Virus Res. 121:11-6.

Shresta, S., Kyle, J.L., Snider, H.M., Basavapatna, M., Beatty, P.R., and Harris, E.

(2004). Interferon-dependent immunity is essential for resistance to primary dengue

virus infection in mice, whereas T- and B-cell-dependent immunity are less critical.

J. Virol. 78: 2701-2710.

Shrestha, B., and Diamond, M.S. (2004). Role of CD8+ T cells in control of West Nile

virus infection. J. Virol. 78: 8312-8321.

Shrestha, B., Gottlieb, D., and Diamond, M.S. (2003). Infection and injury of neurons

by West Nile encephalitis virus. J. Virol. 77: 13202-13213.

Shrestha, B., Samuel, M.A., and Diamond, M.S. (2006). CD8+ T cells require perforin

to clear west nile virus from infected neurons. J. Virol. 80: 119-129.

Shtrichman, R., and Samuel, C.E. (2001). The role of gamma interferon in

antimicrobial immunity. Curr. Opinion. Microbiol. 4: 251-259.

Shueb, R.H. (2002). Abrogation of host resistance in flavivirus resistant mice

REFERENCES

269

following infection with Kunjin virus. MSc. Prelim. Discipline of Microbiology,

School of Biomedical, Biomolecular and Chemical Sciences, The University of

Western Australia.

Shueb, R.H., Pantelic, L. and Urosevic, N. (2005). A delayed morbidity in genetically

resistant mice triggered by some flaviviruses. Arbovir. Res. Australia. 9: 344-351.

Silvia, O.J. (1999). The analysis of natural resistance to flaviviruses in mice and

different cell culture models. Ph.D. thesis. Discipline of Microbiology, School of

Biomedical and Chemical Sciences, The University of Western Australia.

Silvia, O.J., and Urosevic, N. (1999). Variations in LPS responsiveness among

different mouse substrains of C3H lineage and their congenic derivative sublines.

Immunogenetics. 50: 354-357.

Silvia, O.J., Pantelic, L., Mackenzie, J.S., Shellam, G.R., Papadimitriou, J., and

Urosevic, N. (2004). Virus spread, tissue inflammation and antiviral response in brains

of flavivirus susceptible and resistant mice acutely infected with Murray Valley

encephalitis virus. Arch. Virol. 149: 447-464.

Silvia, O.J., Shellam, G.R., and Urosevic, N. (2001). Innate resistance to flavivirus

infection in mice controlled by Flv is nitric oxide-independent. J. Gen. Virol. 82: 603-

607.

Sitati E.M., and Diamond, M.S. (2006). CD4+ T-cell responses are required for

clearance of West Nile virus from the central nervous system.

J Virol. 80:12060-12069.

Sittisombut, N., Maneekam, N., Kanjanahaluethai, A., Kasinrerk, W., Viputtikul, K.,

and Supawadee, J. (1995). Lack of augmenting effect of interferon-gamma on dengue

virus multiplication in human peripheral blood monocytes. J.Med.Virol. 45: 43-49.

Smith, A.L. (1981). Genetic resistance to lethal flavivirus encephalitis: effect of host

age, and immune status and route of inoculation on production of interfering Banzi

virus in vivo. Am. J. Trop. Med. Hyg. 30: 1319-1232.

Smith, P.L., Lombardi, G., and Foster, G.R. (2005). Type I interferon and the innate

immune response – more than just antiviral cytokines. Mol. Immunol. 42: 869-877.

REFERENCES

270

Solomon, T., and Mallewa, M. (2001). Dengue and other emerging flaviviruses. J.

Infect. 42:104-15.

Solomon, T., and Vaughn, DW. (2002). Pathogenesis and clinical features of Japanese

encephalitis and West Nile virus infections. Curr. Topic Microbiol. Immunol. 267:

171-194.

Solomon, T., Dung, N.M., Kneen, R., Gainsborough, M., Vaughn, D.W., and Khanh,

V.T. (2000). Japanese encephalitis. J. Neurol. Neurosurg. Psychiatry. 68: 405-415.

Solomon, T., Dung, N.M., Wills, B., Kneen, R., Gainsborough, M., Diet, T.V., Thuy,

T.T., Loan, H.T., Khank, V.C., Vaughn, D.W., White, N.J., and Farrar, J.J. (2003).

Interfon alpha-2a in Japanese encephalitis: A randomized double-blind placebo-

controlled trial. Lancet. 361: 821-826.

Soos, J.M., Ashley, T.A., Morrow, J., Patarroyo, J.C., Szente, B.E., and Zamvil, S.S.

(1999). Differential expression of B7 co-stimulatory molecules by astrocytes

correlates with T cell activation and cytokine production.

Int Immunol. 11:1169-79.

Sreenivasan, V., Ng, K.L., and Ng, M.L. (1993). Brefeldin A affects West Nile virus

replication in Vero cells but not C6/36 cells. J. Virol. Methods. 45: 1-17.

Steriade, M. and Llines, R. (1988). The functional states of the thalamus and the

associated neuronal interplay. Physiol. Rev. 68: 699-742.

Stohlman, S.A., Bergmann, C.C., Lin, M.T., Cua, D.J., and Hinton, D.R. (1998). CTL

effector function within the central nervous system required CD4+ T cells. J.

Immunol. 160: 2876-2904.

Stohlman, S.A., Bergmann, C.C., Lin, M.T., Cua, D.J., and Hinton, D.R. (1998). CTL

effector function within central nervous system requires CD4+ T cells. J. Immunol.

160: 2896-2904.

Streit, W. (2002). Microglia as neuroprotective, immunocompetent cells of the CNS.

GLIA. 40: 133-139.

Streit, W.J., Mrak, R.E., and Griffin, W.S. (2004). Microglia and neuroinflammation:

REFERENCES

271

a pathological perspective. J Neuroinflammation. 30:1-14.

Sumiyoshi, H., Mori, C., Fuke, I., Morita, K., Kuhara, S., Kondou, J., Nagamatu, H.,

and Igarashi, A. (1987). Complete nucleotide sequence of the Japanese encephalitis

virus genome RNA. Virology. 161: 497-510.

Suzuki, T., Ogata, A., Tashiro, K., Nagashima, K., Tamura, M., Yasui, K., and

Nishihira, J. (2000). Japanese encephalitis virus up-regulates expression of

macrophage migration inhinitory factor (MIF) mRNA in the mouse brain. Biochim.

Biophys. 1517: 100-106.

Takada, K., Masaki, H., Konishi, E., Takahashi, M., and Kurane, I. (2000). Definition

of an epitope on Japanese encephalitis virus (JEV). Arch. Virol.145: 523-534.

Thomsen, A.R., Nansen, A., Madsen, A.N., Bartholdy, C., and Christensen, J.P.

(2003). Regulation of T cell migration during viral infection: role of adhesion

molecules and chemokines. Immunol. Letters. 85: 119-127.

Tsai, T.F. (2000). Flaviviruses. In: Mandell GL, Bennet JE, Bolin R, editors. Volume

2: Infectious Diseases and Their Etiologic Agents, Principles and Practice of

Infectious Diseases. Fifth edition. Philadelphia, Pensylvania: Churchill Livingstone;

2000, p1715-1736.

Tsai, T.F., Popovici, F., Cernescu, C., Campbell, G.L. and Nedelcu, N.I. (1998). West

Nile encephalitis epidemic in southeastern Romania. Lancet. 352: 767-771.

Urosevic, N. (2003). Is flavivirus resistance interferon type I-independent?

Immunol. Cell Biol. 81:224-9.

Urosevic, N., and Shellam, G.R. (2002). Host genetic resistance to Japanese

encephalitis group viruses. Curr Topics Microbiol Immunol, 267: 153-170.

Urosevic, N., Mann, K., Hodgetts, S.I., and Shellam, G.R. (1997b). The use of

microsatellites in high-resolution around the mouse flavivirus resistance locus (Flv).

Arbovirus Res. Aust. 7: 296-299.

Urosevic, N., Ondine, J.S., Sangster, M.Y., Mansfield, J.P., Hodgetts, S.I., and

Shellam, G.R. (1999). Development and characterization of new flavivirus resistant

mouse strains bearing Flvr-like and Flv

mr alleles from wild or wild-derived mice. J.

REFERENCES

272

Gen. Virology. 80: 897-906.

Urosevic, N., Sangster, M.Y., Mansfield, J.P., and Shellam, G.R. (1993). Flavivirus

resistance (Flvr) gene is mice: mapping studies. Arbovirus Res. Aust. 6: 130-134.

Urosevic, N., Silvia, O.J., and Shellam G.R. (2000). Host natural resistance to

flaviviruses controlled by Flv. Res. Adv. Virol. 1: 79-89.

Urosevic, N., van Maanen, M., Mansfield, J.P., Mackenzie, J.S., and Shellam, G.R.

(1990). Molecular characterization of virus-specific RNA produced in the brains of

flavivirus susceptible and resistant mice after challenge with Murray Valley

encephalitis virus. J. Gen. Virol. 78: 23-29.

Vainio, T. (1963a). Virus and hereditary resistance in vitro. I. Behavior of West Nile

(E101) virus in the cultures prepared from genetically resistant and susceptible strains

of mice. Ann. Med. Exp. Biol. Fenn. 41: 1-24.

Vainio, T. (1963b). Virus and hereditary resistance in vitro. II. Behavior of West Nile

(E101) virus in the cultures prepared from challenged resistant, challenged backcross

and non-challenged susceptible mice. Ann. Med. Exp. Biol. Fenn. 41: 25-35.

Vainio, T., Gwatkin, R., and Koprowski, H. (1961). Production of interferon by brains

of genetically resistant and susceptible mice infected with West Nile Virus. Virology.

14: 385-387.

van der Most, R.G., Harrington, L.E., Giuggio, V., Mahar, P.L., and Ahmed, R.

(2002). Yellow fever virus 17D envelope and NS3 proteins are major targets of the

antiviral T cell response in mice. Virology. 296: 117-124.

van Furth R. (1977). The role of phagocytic cells in the defence of the host.

Acta Paediatr Belg. 30: 133-144.

van Furth, R., Diesselhoff-Den Dulk, M.M.C., and Mattie, H. (1973). Quantitative

study on the production and kinetics of mononuclear phagocytes during an acute

inflammatory reaction. J. Exp. Med. 138: 1314-1328.

van Furth, R. (1981). Current view of the mononuclear phagocyte system. Haematol.

Blood Transfus. 27: 3-10.

REFERENCES

273

van Rooijen, N., and Sanders, A. (1994) Liposome mediated depletion of

macrophages: mechanism of action, preparation of liposomes and applications. J

Immunol Methods. 174: 83-93.

Vasconcelos, P.F., Rosa, A.P., Rodrigues, S.G., Rosa, E.S., Monteiro, H.A., Cruz,

A.C., Barros, V.L., Souza, M.R., and Rosa, J.F. (2001). Yellow fever in ParAi State,

Amazon region of Brazil, 1998-1999: entomologic and epidemiologic findings.

Emerg. Infect. Dis. 7: 565-569.

Vilhardt, F. (2005). Microglia: phagocyte and glia cell. Int. J. Biochem. Cell. Biol. 37:

17-21.

Wang, T., Scully, E., Yin, Z., Kim, J.H., Wang, S., Yan, J., Mamula, M., Anderson,

J.F., Craft, J., and Fikrig, E. (2003a). IFN-producing dT cells help control murine

West Nile virus infection. J. Immunol. 171: 2524-2431.

Wang, T., Town, T., Alexopoulou, L., Anderson, J.F., Fikrig, E., and Flavell, R.A.

(2004a). Toll-like receptor 3 mediates West Nile virus entry into the brain causing

lethal encephalitis. Nat. Medicine. 10: 1366-1373

Wang, Y., Lobigs, M., Lee, E., and Mullbacher, A. (2003b). CD8+ T cells mediate

recovery and immunopathology in West Nile virus encephalitis. J. Virol. 77: 13323-

13334.

Wang, Y., Lobigs, M., Lee, E., and Mullbacher, A. (2004b). Exocytosis and Fas

mediated cytolytic mechanisms exert protection from West Nile virus induced

encephalitis in mice. Immunol. Cell Biol. 82: 170-173.

Wang, Y., Lobigs, M., Lee, E., Koskinen, A., and Mullbacher A. (2006). CD8(+) T

cell-mediated immune responses in West Nile virus (Sarafend strain) encephalitis are

independent of gamma interferon. J Gen Virol. 87:3599-609.

Webster, L.T. (1937). Inheritence of resistance of mice to enteric bacterial and

neurotropic viral infections. J. Exp. Med. 65: 261-286.

Webster, L.T., and Clow, A.D. (1936). Experimental encephalitis (St. Louis type) in

mice with high inborn resistance. A chronic subclinical infection. J Exp. Med. 63:

827-945.

REFERENCES

274

Webster, L.T., and Johnsen, M.S. (1941). Comparative virulence of St. Louis

encephalitis virus cultured with brain tissue from innately susceptible and innately

resistant mice. J. Exp. Med. 74: 489-494.

Westaway, E.G. (1987). Flavivirus replication strategy. Adv Virus Res. 33: 45-90.

Westaway, E.G., Brinton, M.A., Gaidamovich, S.Y, Horzinek, M.C., Igarashi, A.,

Kaariainen, L., Lvov, D.K., Porterfield, J.S., Russell, P.K., and Trent, D.W. (1985).

Flaviviridae: Report of Togaviridae study group, Vertebrate Virus Subcommittee,

International Committee on Taxonomy of Viruses. Intervirology. 24: 183-192.

Westaway, E.G., Mackenzie, J.M., and Khromykh, A.A. (2002). Replication and gene

function in Kunjin virus. Curr. Topic. Microbiol. Immunol. 267: 323-352.

Westaway, E.G., Mackenzie, J.M., Kenney, M.T., Jones, M.J., and Khromykh, A.A.

(1997). Ultrasutructure of Kunjin virus infected cells: colocalization of NS1 and NS3

with double-stranded RNA and of NS2B with NS3 in virus induced membrane

structures. J. Virol. 71: 6650-6661.

Whitmire, J.K., Tan, J.T., and Whitton, J.L. (2005). Interferon-γ acts directly on CD8+

T cells to increase their abundance during virus infection. J. Exp. Medicine. 201:

1053-1059.

Whitton, J.L., Slifka, M.K., Liu, F., Nussbaum, A.K., and Whitmire JK. (2004). The

regulation and maturation of antiviral immune responses.

Adv Virus Res. 63:181-238.

Winter, P.M., Dung, N.M., Loan, H.T., Kneen, R., Wills, B., Thu, L.T., House, D.,

White, N.J., Farrar, J.J., Hart, A., Solomon, T. (2004). Proinflammatory cytokines and

chemokines in humans with Japanese encephalitis. J. Infect. Dis. 190: 1618-1626.

Wu, G.F., and Perlman, S. (1999). Macrophage infiltration, but not apoptosis is

correlated with immune-mediated demyelination following murine infection with a

neurotropic coronavirus. J. Virol. 73: 8771-8780.

Wu, J., Bera, A.K., Kuhn, R.J., and Smith, J.L. (2005). Structure of the Flavivirus

helicase: Implications for catalytic activity, protein interactions, and proteolytic

processing. J. Virol. 79: 10268-10277.

REFERENCES

275

Wu, S.J., Grouard-Vogel, G., Sun, W., Mascola, J.R., Brachtel, E., Putvatana, R.,

Louder, M.K., Filgueira, L., Marovich, M.A., Wong, H.K., Blauvelt, A., Murphy,

G.S., Robb, M.L., Innes, B.L., Birx, D.L., Hayes, C.G., and Frankel, S.S. (2000).

Human skin Langerhans cells are targets of dengue virus infection. Nature Med. 6:

816-820.

Xiao, S.Y., Guzman, H., Zhang, H., Travassos da Rosa, A.P., and Tesh, R.B. (2001).

West Nile virus infection in the golden hamster (Mesocricetus auratus): a model for

West Nile encephalitis.Emerg Infect Dis. 7:714-21.

Yamshchikov, V.F., and Compans, R.W. (1993). Regulation of the late events in

flavivirus protein processing and maturation. Virology. 192: 38-51.

Yang, D., Yeh, W.T., Chen, R.F., Chuon, H.L., Tsai, H.P., Yao, C.W., and Shaio,

M.F. (2004). A model to study neurotropism and persistency of Japanese encephalitis

virus infection in human neuroblastoma cells and leukocytes. J. Gen. Virol. 85: 635-

642.

You, S., Falgout, B., Markoff, L., and Padmanabhan, R. (2001). In vitro RNA

synthesis from exogenous dengue viral RNA templates requires long range

interactions between 5'- and 3'-terminal regions that influence RNA structure. J Biol

Chem. 276: 15581-15591.

Zhao, J., Hinton, D.R., Stohlman, S.A., Liu, C.P., Zhong, L., and Marten, N.W.

(2005). Maintenance of CD8+ T cells during acute viral infection of the central

nervous system required CD4+ T cells but not Interleukin-2. Virol. Immunol. 18: 162-

169.

Zhou, A., B. A. Hassel, and R. H. Silverman. 1993. Expression cloning of 2-5A-

dependent RNase: a uniquely regulated mediator of interferon action. Cell 72:753-

765.

Zisman, B., Wheelock, E.F., and Allison, A.C. (1971). Role of macrophages and

antibody in resistance of mice against yellow fever virus. J Immunol. 107: 236-43.