in-vitro characterization of human brain

120
IN-VITRO CHARACTERIZATION OF HUMAN BRAIN MICROVASCULAR ENDOTHELIAL CELLS INFECTED WITH WEST NILE VIRUS TO STUDY TIGHT JUNCTION INTEGRITY A THESIS SUBMITTED TO THE GRADUATE DMSION OF THE UNIVERSITY OF HAWAI'IIN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE IN BIOMEDICAL SCIENCES (TROPICAL MEDICINE) MAY 2008 By Yeung Y. Lo Thesis Committee: VlVek R. Nerurkar, Chairperson Saguna Verma Frederic Mercier

Upload: others

Post on 22-Dec-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

IN-VITRO CHARACTERIZATION OF HUMAN BRAIN

MICROVASCULAR ENDOTHELIAL CELLS INFECTED WITH WEST

NILE VIRUS TO STUDY TIGHT JUNCTION INTEGRITY

A THESIS SUBMITTED TO THE GRADUATE DMSION OF THE

UNIVERSITY OF HAWAI'IIN PARTIAL FULFILLMENT OF THE

REQUIREMENTS FOR THE DEGREE OF

MASTER OF SCIENCE

IN

BIOMEDICAL SCIENCES (TROPICAL MEDICINE)

MAY 2008

By Yeung Y. Lo

Thesis Committee:

VlVek R. Nerurkar, Chairperson Saguna Verma

Frederic Mercier

We certify that we have read this thesis and that, In our opinion, It Is

satisfactory In scope and quality as thesis for the degree of Master of

Science In Biomedical Sciences (Tropical Medicine).

THESIS COMMITTEE:

Chairperson

ii

Dedications

I would like to dedicate this work to my loved ones who have given me their love

and support unconditionally and will motivate me for many more challenges to

come.

Tao Chih Lo

Lai Fong Kong

Sin Sin Lo

Ning Ning Lo

FaiLo

Aja Merkel Razumny

Roxanne Menade/ook

AnahShah

Robert Mourant

Louise Mourant

Michael TutteffOw

iii

Acknowledgments

I thank my committee members and the funding sources for making this project

possible. I would like to give special thanks to Dr. Saguna Verma for her day-to­

day guidance, from the beginning to the end of this project, and beyond. I also

appreciate her patience and support throughout my time in the laboratory. My

funding sources included grants from the Centers for Biomedical Research

Excellence (P20RR018727), Research Centers in Minority Institutions Program

(G12RR003061), NCRR, NIH; and Hawaii Community Foundation (20050405).

In addition, I would like to dedicate my sincere appreciation to my fellow students

and co-workers who have helped me in many different ways to make this project

come through. Special thanks go to Ulziijargal Gurjav, Laami Submibcay, James

Kelley, Haiyan Luo, Stephanie Lum, Austin Nakatsuka, Pakieli Kaufusi, Moti

Chapagain, Becky Nakama and Juliene Co.

iv

Abstract

West Nile virus (WNV) is the predominant cause of viral encephalitis in the

United States today. Infection of neurons and induction of inflammatory

response are cardinal features of fatal WN encephalitis. WNV neuroinvasion has

been implicated to occur by hematogenous spread and possibly via the blood­

brain barrier (BBB) interface. However, the precise mechanisms of viral entry

into the CNS through the brain vascular endothelium, key component of the BBB,

are unclear. Cellular and molecular modulations of tight junction proteins (T JP),

cell adhesion molecules (CAM) and matrix metalloproteinases (MMP) on brain

vascular endothelial cells in response to WNV infection were highly likely to

influence transmigration of WNV into the CNS. We hypothesized that WNV

could infect and replicate in human brain microvascular endothelial (HBMVE)

cells. Further we hypothesized that infection with WNV would lead to modulation

in the expressions of TJP, CAM and MMP, thereby leading to enhanced

migration of the virus across the BBB. Our results demonstrate that WNV could

efficiently replicate in HBMVE cells. In correspondence to the maximal viral

replication and virion release on day 2 after infection, WNV claudin-1

transcription and translation were significantly up-regulated. In addition,

significant induction of E-selectin and VCAM-1 transcription was detected.

VCAM-1 increase was further confirmed at post-translational level. WNV

selectively modulated the expressions of CAM and TJP, but none of the MMP

examined. These modulations are confirmed to be direct effects of WNV

replication as changes in CAM or T JP expressions were not observed in HBMVE

v

cells infected with UV-inactivated WNV. Our in vitro results suggest an active

role of brain endothelial cells in WNV neuroinvasion.

vi

Table of Contents

Contents Page

Dedications ...................................................................................... iii

Acknowledgements .......................................................................... .iv

Abstract ........................................................................................... v

Table of Contents .............................................................................. vii

List of Tables ...... '" ........................................................................... x

List of Figures ......................................................... '" ....................... xi

List of Abbreviations and Symbols ......................................................... xii

Chapter 1. Introduction

1.1 West Nile Virus and Human Diseases ........................................... 1

1.1.1 West Nile Virus ................................................................ 1

1.1.2 Epidemiology of WN Diseases ............................................ 4

1.1.3 Acquisition and Dissemination of WNV in Humans .................. 6

1.1.4 InfiammatoryWNNDand Pathology ..................................... 7

1.1.5 Immune Response to WNV Infection ................................... 1 0

1.1.6 Diagnosis. Treatment and Prognosis ................................... 14

1.1.7 Vaccines and Disease Prevention ....................................... 15

1.2 Blood-Brain Barrier and Viral Infection .......................................... 17

1.2.1 Introduction to BBB and Transmigration Pathways

Through BBB .................................................................. 17

1.2.2 Dysfunction of BBB in Viral Infections ................................... 20

1.2.2.1 T JP and BBB Permeability ................................ 20

1.2.2.2 MMP and TJP ................................................ 24

1.2.2.3 CAM and Immune Cell Transmigration ................ 25

vii

Chapter 2. Thesis Scope

2.1 Background for Research Question: Whether HBMVE cells play

a role in WNV-CNS entry? ................................................................... 32

2.2 Objectives and Hypothesis ........................................................ 32

2.3 Specific Aims ......................................................................... 33

2.4 Significance ............................................................................ 33

Chapter 3. Methods

3.1 Experimental Design for Specific Aim 1 ........................................ 34

3.1.1 Infection of HBMVE cells in 6-well plates and

on cover slips ................................................................ 34

3.1.2 Collection of culture supernatants, harvesting of cells

and fixation of cells on cover slips ...................................... 34

3.1.3 Detection of WNV on cover slips by immunofluorescence ....... 36

3.1.4 Quantitation of virus titer in supernatants by Plaque assay ...... 36

3.1.5 Quantitation of virus RNA copy in supernatant by qRT -PCR. ... 37

3.1.5.1 Extraction of viral RNA from supernatants ......... 38

3.1.5.2 Synthesis of cDNA from viral RNA

from supernatants ........................................ 38

3.1.6 Quantitation of virus RNA copy in infected HBMVE cells

by qRT-PCR ................................................................. 38

3.1.6.1 Extraction of cellular RNA from celllysates ........ 39

3.1.6.2 Synthesis of cDNA from cellular RNA ............... 39

3.2 Experimental Design for Specific Aim 2 ...................................... .40

3.2.1 Infection of HBMVE cells in 6-well plates, T-25 tissue

culture flasks and on cover slips ........................................ 40

3.2.2 Harvesting of cells and fixation of cells on cover slips ........... .40

3.2.3 Evaluation of transcription fold-change of genes

of interest by qRT-PCR. ................................................. .40

3.2.4 Detection of the expression of proteins of interest of

viii

bywestem blot.. ................................................ : ........... 41

3.2.5 Detection of proteins of interest on cover slips by

immunofluorescence ...................................................... .41

3.3 Materials ............................................................................... 43

3.3.1 HBMVE cells ................................................................ .43

3.3.2 WN virus ...................................................................... 43

3.3.3 Antibodies ..................... '" ...... '" ................................... 44

Chapter 4. Results

4.1 WNV infected and replicated in HBMVE cells ................................ 45

4.2 Replication-competent WNV, but not W-inactivated WNV,

differentially up-regulated T JP expressions in HBMVE cells ............. 54

4.3 Replication-competent WNV, but not W-inactivated WNV,

selectively induced CAM expressions in HBMVE cells .................... 59

4.4 Infection of HBMVE cells with WNV did not result in

MMP or TIMP expressions ........................................................ 64

Chapter 5. Discussion ................................................................... 65

References ..................................................................................... 72

ix

Tables

Table 1

Table 2

List of Tables

Page

Vaccines licensed or in clinical trials .................................... 17

Percent ofWNV-infected cells by IFS staining ....................... 46

x

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

Fig. 6

Fig. 7

Fig. 8

Fig. 9

Fig. 10

List of Figures

Page

Schematic of WNV genome ............................................... 2

Schematic of WNV intracellular life cycle .............................. 3

Proposed interactions of junctional complex proteins ............. 23

Adhesion receptors involved in the diapedesis process .......... 29

WNV could infect and replicate in HBMVE cells .................... 46

WNV titer recovered from cell culture supematant quantitated by plaque assay ............................................ .48

WNV transcripts recovered from cell culture Supematant quantitated by qRT-PCR. ................................ 50

Intracellular WNV RNA transcripts quantitated by qRT-PCR .................................................................. 52

WNV differentially modulated T JP expressions quantitated by qRT- PCR, we and IFS ............................... 56

WNV selectively induced CAM expression quantitated by qRT-PCR, WB and IFS ................................. 61

xi

(k)Da

(s)PECAM

I-Ig

I-Im

ACBRI

AFP

AP

B(M)VEC

BBB

BSA

C

CAM

CCl-12

CCR5(l132)

CDC

cDNA

CMV

CNS

C02

CSF

D

DC-SIGN

ddH20

DEET

DNA

DNase

E

ECl

ECM

List of Abbreviations and Symbols

kilodalton

soluble platelet endothelial cell adhesion molecule

microgram

micron

Applied Cell Biology Research Institute

acute flaccid paralysis

alkaline phosphatase

brain (micro)vascular endothelial cells

blood-brain barrier

bovine serum albumin

capsid

cell adhesion molecule

Chemokine (C-C motif) ligand 2

chemokine (C-C motif) receptor 5 delta 32

Center for Disease Control and Prevention

complementary deoxyribonucleic acid

cytomegalovirus

central nervous system

carbon dioxide

cerebral-spinal fluid

day

dendritic-cell-specific, ICAM-3-grabbing non-integrin

double-distilled water

N,N'-Diethyl-m-toluamide

deoxyribonucleic acid

deoxyribonuclease

envelope

enhanced chemiluminescence

extracellular matrix

xii

EGF

EHV-1

h

HAD

HBMVE

HBV

HCMV

HIV

HSV

ICAM

IFN

IFS

IgMlG

IL

IP-10

IRF-3

JAM

MDCK

MHC

MIG

min

mL

MMP

MOl

MV

NeuroAIDS

NF-KB

NK

NMDA

NS

NY99

epidermal growth factor

equine herpes virus-1

hour

HIV-associated dementia

human brain microvascular endothelial

hepatitis B virus

human cytomegalovirus

human immunodeficiency virus

herpes simplex virus

intercellular adhesion molecule

interferon

immunofluorescence staining

immunoglobulin M or G

interleukin

Chemokine (C-X-C motif) ligand 10

Interferon regulatory factor 3

junctional adhesion molecule

Madin-Darby canine kidney

major histocompatibility complex

monokine induced by gamma interferon

minute

milliliter

matrix metalloproteinase

multiplicity of infection

measles virus

neuro aquired immunodeficiency syndrome

nuclear factor-kappa B

natural killer

N-methyl D-aspartate

nonstructural

New York 99

xiii

°C PBS PCR

PFA PFU prM

PVDF qRT-PCR

RNA rpm

RT

50 50S-PAGE

slV

TBs(T)

TIMP

TJP

TLR

TMEV

TNF

UV

VCAM

we WNF

WNNO

WNV

WNVE

degree Celsius

phosphate buffer saline

polymerase chain reaction

paraformaldehyde

plaque-forming unit

pre-membrane

polyvinylidene fluoride

quantitative reverse-transcriptase polymerase chain

reaction

ribonucleic acid

revolutions per minute

room temperature

standard deviation

sodium dodecyl sulfate polyacrylamide gel

electrophoresis

simian immunodeficiency virus

tris-bufferred saline (Tween)

tissue inhibitor of metalloproteinase

tight junction protein

toll-like receptor

Theiler's murine enceaphalomyelitis virus

tumor necrosis factor

ultraviolet

vascular cell adhesion molecule

western blot

West Nile fever

West Nile neurological disease

West Nile virus

Wes Nile virus encephalitis

xiv

Chapter 1. Introduction

1.1 West Nile virus and human diseases

1.1.1 West Nile virus

West Nile virus (WNV) is an arthropod-borne virus transmitted by mosquito

vectors first isolated from a febrile woman in the West Nile region of Uganda in

1937 (Smithbum K, 1940). It belongs to the Japanese encephalitis serocomplex

of the genus Flavivirus within the family of Flaviviridae (Brinton, 2002). WN

virions are spherical in shape with icosahedral symmetry. Its envelope measures

50 nm in diameter and it has a smooth spike-less outer surface (Mukhopadhyay

et a/., 2003).

WNV is a single-stranded positive-sense RNA virus of approximately 10.8 kb

genome size consisting of three structural proteins and seven non-structural (NS)

proteins. Structural proteins include (i) capsid (C) protein which is responsible for

viral RNA binding, (ii) membrane (prM) protein that blocks premature fusion and

chaperone envelope (E)-protein folding, and (iii) envelope (E) which mediates

viral attachment, and membrane fusion and viral assembly (Mukhopadhyay et a/.,

2005). The seven NS proteins are NS1, NS2A, NS2B, NS3, NS4A, NS4B and

NS5 (see Fig.1) (Mukhopadhyay et a/., 2003). The viral NS proteins regulate

viral transcription, replication and attenuate host antiviral responses (Samuel &

Diamond, 2006). The WNV genome is contained within a capsid, which is

surrounded by a host-derived lipid membrane embedded with virus-encoded E

and prM glycoproteins (Mukhopadhyay et aI., 2003). The E protein contains

epitopes recognizable by neutralizing antibodies and is responsible for receptor

binding and fusion with target cell membrane (Brinton, 2002; Chu et a/., 2005).

Structura l NCR NCR NCR

5{) )) )) C-prM-E NS1-NS2A-NS2B-NS3-NS4A-NS4B-NS5

POIYP~ ~o- 1 1 and post-translational modification Serine protease RNA polymerase

NTPase RNA helicase

Fig.1 Schematic of WNV genome [ Mukhopadhyay S. et aI., Nat. Rev. Microbial., 2005,3(1 ):13-22]

The linear positive-sense genome serves directly as messenger RNA with a

single open reading frame containing the necessary RNA-dependent RNA

polymerase, encoded by NS proteins, for replication (Brinton, 2002).

Immunoglobulin-like Domain III of WNV envelope protein is responsible for

binding to receptor on the surface of Vero and C6/36 mosquito host cells (Chu et

a/., 2005). While WNV receptor on neurons is unclear, a putative WNV receptor,

integrin av133, has been identified on non-neuronal cells such as Vero and CS-1

melanoma cells (Chu & Ng, 2004b; Lee et a/., 2006). Other molecules which

have been implicated in the attachment of WNV to cells in vitro include DC-SIGN

and DC-SIGN-R (Davis et aI., 2006a).

2

WNV enters host cells via clathrin-mediated endocytic pathway where virus is

transported toward the endoplasmic reticulum (ER) via lysosomal vesicles (Chu

& Ng, 2004a). At low pH, the viral envelope fuses with the lysosomal membrane

and viral RNA is subsequently released into the cytoplasm. Virus replication

occurs in the cytoplasm in close proximity to rough ER. Assembly of virions

takes place within the ER lumen, and the nascent virions are transported within

vesicles to the cell surface and released by exocytosis (Brinton, 2002). See Fig.

2.

.. RIlla CD'" 8IIIf/IJIg Md _-_...... PnIf8III rlllJ&' II'OLI

",,-.. - "­...... - .. ,.. .-. II"-........ - .. ~ ...... - .. 1

Fig. 2 Schematic of WNV Intracellular life cycle. WNV attaches to susceptible cells through an as yet uncharacterized cell surface receptor and enters cells via receptor-mediated endocytosis. After acidification of the endosome, the E protein undergoes a conformational change that facilitates membrane fusion and nucleocapsid escape into the cytoplasm. Viral RNA binds to ribosomes in the cytoplasm and Is recruited to the rough endoplasmic reticulum where translation of the polyprotein from a single open reading frame ensues. The polyprotein is cleaved post-translationally by viral and host proteases. Translation, replication, and packaging are coupled processes and nascent viruses accumulate in membrane-derived vesicles prior to secretion by the cellular exocytosis machinery. [Diamond MS et ai, Viral Immuno/., 2003, 16(3):259-78]

3

1.1.2 Epidemiology of WN diseases

Since 1937, WNV has caused epidemics of febrile illnesses and sporadic

encephalitis throughout Africa, Europe, Asia and Australia (Hayes & Gubler,

2006; Murgue et al., 2002). In 1990s, fatal WNV encephalitis started to

frequently manifest in epidemics in Romania, Russia, and Israel. In 1999, WNV

infection was documented the first time in North America in New York City,

causing 59 cases of encephalitis and 7 deaths (Hayes & Gubler, 2006; Murgue et

al., 2002). Nucleotide sequencing of the envelope gene from isolated WNV

strain New York 99 (NY99) was analyzed and it was revealed to be closely

related (>99.8% amino acid homology) to an Israeli goose isolate in 1998

(Lanciotti et al., 1999). After its introduction to the U.S. in 1999, the virus has

spread dramatically westward across the United States, southward into Central

America and the Caribbean, and northward into Canada, resulting in the largest

epidemics of neuroinvasive WNV disease ever reported (Hayes et al., 2005a).

Specifically, the U.S. epidemic in 2003 was the largest WNV encephalitis

outbreak ever reported (Solomon, 2004). WNV is divided into lineages 1 and 2

based on genetic sequencing (Hayes & Gubler, 2006). Lineage 1 includes

pathogenic strains from North America, Europe, Australia, Africa, and Asia,

whereas lineage 2 includes only the less virulent strains from Africa (including the

original Uganda strain) and Madagascar (Lanciotti et al., 2002). Among lineage

1, WNV is categorized into four clades based on unique amino acids

polymorph isms or deletions in their envelope proteins (Brinton, 2002; Kuno et al.,

4

1998): Indian, Kunjin, A, and B (Beasley at al., 2004). Isolates from the US are in

clade B of lineage 1 and are closely related to strains from Israel.

WNV is enzootically maintained between more than 200 species of birds and

many species of mosquitoes (van der Meulen at al., 2005). In the U.S., different

species of mosquitoes play roles in WNV transmission in different parts of the

U.S. depending on the geographical location: Culax tarsalis and quinquafasciatus

in the West, Culax quinquafasciatus, and nigripalpus in the Southeast, and Culax

pipians and restuans in the Northeast. Mosquito acquires WNV from infected

blood through a blood meal, after penetrating the gut. WNV replicates in the

nervous system and salivary gland and persists throughout the life of the

mosquito without producing disease (Girard et al., 2005). Most birds support

high-titer of WNV replication in the blood, some can last for up to 100 days

without developing fatal illnesses, therefore the amplifying hosts (van der Meulen

at al., 2005). In naturally infected crows and blue jays, WNV has been detected

in the brains, livers, lungs, spleens, hearts and kidneys (Samuel & Diamond,

2006). Many other vertebrates are also susceptible to WNV infection but

neurological disease is only common in horses (Kleiboeker at al., 2004). It has

been suggested that the initial WNV introduction into New York city came from

infected migratory birds (Rappole at al., 2000), or imported exotic birds. Humans

can also contract WNV by means other than mosquito bites though at significant

lower probability. The less common routes of human WNV transmission include

transfusion of WNV-infected blood products (Kleinman at al., 2005; Pealer at al.,

5

2003; Stramer et aI., 2005), transplantation of WNV-infected organs (Bragin­

Sanchez & Chang, 2005; Cushing et al., 2004; Wadei et al., 2004), vertical

mother-to-child transplacental spread (2002a), breast feeding of milk from WNV­

infected mother (2002c) and accidental laboratory exposure (2002b).

Multiple epidemiological data suggests people with compromised immune

systems, including the AIDS and transplant patients (Guamer et a/., 2004), and

older age, being male, having underlying condition of hypertension, or diabetes

mellitus are among the high risk group to develop severe WNV diseases (Jean

CM, ; Murray et al., 2006). In addition, retrospective chart reviews of 172 WNV

cases hospitalized in Houston, Texas, between 2002 and 2004 indicates that

being African-American, being positive for hepatitiS 'C virus and having chronic

renal disease are risk factors for death after age adjustment (Murray et al., 2006).

Further, an underlying genetic risk factor is identified to be homozygosity for

defective CCR5 allele (CCR51132) in a cohort of WNV-infected Caucasian

patients from Arizona and Colorado (Glass at al., 2006). CCR5, critical

chemokine receptor, has been demonstrated to regulate leukocyte trafficking into

the brain for WNV clearance and is crucial for survival in mice infected with WNV

(Glass at al., 2005).

1.1.3 Acquisition and dissemination of WNV in humans

Humans acquire WNV intradermally from the saliva of infected mosquito upon

feeding. WNV is believed to be initially taken up by the skin Langerhans cells

6

(Chambers & Diamond, 2003), which then travel to and replicate in regional

draining lymph nodes where the virus enters the bloodstream for the first time

leading to primary viremia (Johnston et sl., 2000). Secondary viremia follows at

significantly higher titers as a result of WNV dissemination and replication in both

the primary and secondary lymphoid organs (Chambers & Diamond, 2003; Davis

et al., 2006b; Samuel & Diamond, 2006). Virus can be detected in blood as early

as 1 to 2 days after initial inoculation, and viremia typically lasts for a week

(Davis et al., 2006b). Common self-limiting clinical features of WNF include the

abrupt onset of fever, headache, and fatigue, with variable malaise, anorexia,

nausea, myalgia, lymphadenopathy, and a nonpruritic generalized

maculopapular rash (Campbell et al., 2002; Ferguson et al., 2005; Tilley et al.,

2007). Further, through un-identified mechanisms, WNV enters the CNS and

causes an array of West Nile neurological disease (WNND) including meningitiS,

encephalitis, and acute flaccid paralysis/polio-like myelitis (AFP) (Bode et sl.,

2006; Weiss et sl., 2001).

1.1.4 Inflammatory WNND and pathology

Severe outcomes of WN infection are the development of WNND. Postmortem

histologic examinations of WNV encephalitic brains demonstrate perivascular

inflammation, microglial nodules, variable necrosis, and loss of neurons (Guamer

et al., 2004; Kleinschmidt-DeMasters et aI., 2004). Among the CNS, the deep

gray nuclei, brainstem, and spinal cord show most pathology (Guamer et sl.,

2004; Kleinschmidt-DeMasters et sl., 2004). Microglial nodules, loss of anterior

7

hom cells and parivascular infiltration of T- and B-Iymphocytes in the spinal cord

is observed in patients with AFP (Kelley st a/., 2003; Kleinschmidt-DeMasters at

a/., 2004). Immunohistochemistry revealed that WNV mostly being localized

within neurons but also in glia (van Marie st a/., 2007) primarily in the brain stem

and anterior horns. WNV antigen staining is generally focal and sparse.

However, virus can be detected throughout the eNS and in other systemic

organs in immunosuppressed patients (Armah st a/., 2007; Guamer st a/., 2004).

Experimental eNS pathology studies of WNV infection in animal models also

report similar findings as in patients with WNVE, such as infection and injury of

brain stem, hippocampal, and spinal cord neurons but rarely in non-neuronal

tissues (Samuel & Diamond, 2006). Sequential histopathological events

unraveled in a mouse model with WNVE demonstrates that peripheral infection

and replication in the skin, spleen and kidney preceded infection of the eNS

(Garcia-Tapia et a/., 2007).

Acute or chronic neuroinflammation can result in various degree of BBB

breakdown and allow enhanced infiltration of circulating immune cells into the

eNS (Aktas st a/., 2007). Activation of glial cells is increasingly being recognized

to be important in the disease process (Aktas st a/., 2007). Microglia, the resident

immune cells of the brain, as well as astrocytes are capable of releasing a

diverse array of soluble factors, such as proinflammatory cytokines and free

radicals when activated by pathogens such as HIV-1, TNF-a and IL-1(3 are

secreted by HIV-1-infected and activated macrophages as well as microglia and

8

play critical role in HIV-associated dementia, which is characterized by leukocyte

infiltration, microglia activation, aberrant chemokine expression, blood-brain

barrier (BBB) disruption, and ultimately damage and death of neurons (Eugenin

at a/., 2006b). Further, excitatory neurotransmitter such as glutamate can act as

a neurotoxin if it is produced in excess or is not re-uptaken properly by astrocytes

(Tilleux & Hermans, 2007). As seen in NeuroAIDS, TNF-a and IL-113

synergistically over stimulate glutamate receptor N-methyl-D-aspartate (NMDA)

and leads to neuronal cell death by increased calcium influx (Brabers & Nottet,

2006).

In WN meningoencephalitis, infiltration of lymphocytes, perivascular

accumulation of macrophages and microglial nodules (Hayes & Gubler, 2006;

Hayes st a/., 2005b) are grossly observed in biopsies of diseased individuals.

Molecular pro inflammatory parameters such as cytokines and chemokines, MCP-

5 (or CCl12), IP-10 (or CXCl10), and MIG (or CXCl9), IFN-y and TNF-a

(Garcia-Tapia st a/., 2007), are found to be specifically triggered in experimental

WNV CNS infections It is recently shown that the expression of the WNV-NY99

capsid protein by implantation into the striatum of rats results in

neuroinflammation, together with induction of CXCl10 and diminished

expression of the protective astrocyte-specific endoplasmic reticulum stress

sensor" gene (OASIS), which is consistent with that observed in neurons and

astrocytes transfected with the same WNV capsid (van MarIe st a/., 2007). On

similar note, in vitro experimental infection of different human brain cell

9

populations confirmed that both neurons, and to a lesser extent glial cells are

susceptible to WNV infection and support replication at various degrees

(Cheeran at a/., 2005). Despite the borderline low level of WNV infection and

replication in astrocytes and microglia, robust immune mediators are released

including proinflammatory cytokines (ll-6, TNF-a), and chemokines (CXCl10,

CCL2, CCl5) which playa critical role in the recruitment of virus-specific T cells

into the CNS (Glass at a/., 2005; Klein at a/., 2005). In addition to

proinflammatory cytokines, the levels of a critical mediator of the inflammatory

cascade, macrophage migration inhibitory factor (MIF) is found to be increased in

the plasma and CSF of WNV-infected patients (Aljona at al., 2007a). Increased

MIF expression facilitates WNV neuroinvasion by compromising BBB integrity

and is associated with increased mortality in a mouse model (Aljona at al.,

2007a). Elevated levels of MIF has also been suggested to play roles in viral

pathogenesis, including dengue hemorrhagic fever (Chan at aI" 2006) and HBV

in patients (Zhang at al., 2002) and HCMV (Bacher at a/., 2002) and influenza

virus in vitro (Arndt at al., 2002).

1.1.5 Immune response to WNV Infection

Innate and adaptive immunity are both crucial in controlling WNV infection

(Diamond at a/., 2003a). Antiviral type I interferon (IFN-a and (3) production

mediated by RIG-I and MDA5 is essential in suppressing viral titers in the brain

and peripheral organs and is directly associated with reduced lethality in mice

(Fredericksen at al., 2008). In vitro treatment of cells, including primary neurons,

10

with IFN-a~ before or after WNV infection reduces viral titer and inhibits

cytopathology (Samuel & Diamond, 2005). Activation of the complement system

is required for the induction of protective antibodies against WNV, specifically

C1q, C3, C4, factor B, factor D and complement receptors 1 and 2 are critical in

controlling WNV infection in mice (Samuel & Diamond, 2005). Moreover,

altemative complement pathway is important for CD8+ T cell recruitment

whereas classical and lectin pathways are important in modulating both Band T

cells responses to WNV infection (Mehlhop at al., 2005). Toll-like receptor 3

(TLR-3), another pattem recognition receptor that recognizes intracellular dsRNA

to mediate type I interferon production via NF-K8 (Alexopoulou at al., 2001), has

been shown to trigger TNF-a signaling pathway, which leads to augmentation of

WNV entry into the brain to cause lethal encephalitis in mouse models (Wang at

al., 2004). Both humoral (Chung at al., 2007; Diamond at al., 2003b; Engle &

Diamond, 2003; Mehlhop & Diamond, 2008; Nybakken at al., 2005) and cell­

mediated immunity (Brien at al., 2007; Klein at al., 2005; Shrestha & Diamond,

2004; Sitati at al., 2007; Wang at al., 2003a; Wang at aI., 2003b) are important in

WNV clearance. Early induction of neutralizing IgM is particularly crucial in

limiting viremia and WNV dissemination into tha brain which protects against

lethality. Anti-WNV IgM induction later also plays role in anti-WNV IgG response

(Diamond at al., 2003b). Passive transfer of WNV antibody prior to viral

challenge has been shown to confer protection in immunocompetent mice,

however humoral immunity is insufficient in rescuing RAG1 knockout mice which

lack function T and B cells from WNV infection (Diamond et al., 2003a; Engle &

11

Diamond, 2003). One mechanism of clearance of WNV-infected cells is by Fc­

gamma receptor 1- and/or IV-mediated phagocytosis through monoclonal IgG

antibodies recognition of cell surface-associated NS1 (Chung at a/., 2007). Both

CD 4+ (Sitati & Diamond, 2006), CD 8+ cytotoxic T cells (Brien at a/., 2007;

Klein at a/., 2005; Shrestha & Diamond, 2004; Shrestha at a/., 2006; Sitati at a/.,

2007; Wang at a/., 2003b) and IFN-y-producing gamma delta (y6) T cells (Wang

at aI., 2003a) play roles in WNV infection control. In wild-type mice, yi5-T cells

expanded significantly during WNV infection and produced IFN-y. Adoptive

transfer of IFN-y-producing yi5-T cells to knockout mice increase survival of

infected mice (Wang at aI., 2003a). While IgM appears to be crucial factor to

restrict viremia, animals lacking CD8+ T cells or MHC class la have functional

humoral response compared to animal which have CD8+ T cell but bear higher

viral loads in the CNS have higher mortality rates (Shrestha & Diamond, 2004;

Wang at a/., 2003b). Adoptive transfer of naive, unprimed splenic CD8+ T cells to

RAG-KO animals (which lack mature T and B cells) appear to be sufficient in

providing protection against lethal dose WNV infection (Engle & Diamond, 2003).

On the other hand, CD8+ cytotoxic T cells directed against specific WNV antigen

peptides have also been shown to carry out clearance (Brien at a/., 2007; Purtha

at a/., 2007). A study demonstrated that one way CD8+ T cells cleared WNV

from infected neurons is mediated by perf orin pathways (Shrestha at a/., 2006).

Migration of activated CD8+ T cells into the brain is important for viral clearance

as a mouse model that lack CD8+ T cells or major histocompatibility complex

12

class la (MHC-I) antigens, antigen peptide presenting complex to CD8+ T cells,

shows up to 1,000-fold higher titers of WNV in the CNS and increased mortality

after WNV inoculation (Shrestha & Diamond, 2004). CD4+ T cells also

participate in the WNV immunity in promoting the maturation of antibody

response but also in facilitating CD8+ T cells trafficking into the brain by binding

of CD40 and CD40 ligand (CD40l) on activated CD4+ T cells (Sitati et a/., 2007).

Further, for cell-mediated immunity to function optimally, proper immune cells'

trafficking is necessary. Of note, infiltration of activated CD4+ and CD8+ T cells,

NK cells and macrophages relies on chemokine production by infected tissues,

such as CXCl10 by neurons and the receptors CXCR3 (Klein et a/., 2005), as

well as the expression of chemokine receptors and CCR5 and its ligand CCl5

(Glass et a/., 2005).

Immune evasion mechanism has been described in vitro experiments with WNV.

Specifically, WNV envelope protein (WNV-E), independent of TlR-3, blocks the

production of antiviral and poly (I:C)-induced proinfiammatory cytokines TNF-a,

Il-6, and IFN-(3 in murine macrophages at the level of receptor-interacting protein

1(RIP1) to mediate the activation of NF-KB and IFN regulatory factors (lRF)

(A~ona et al., 2007b). Another study using WNV replicon suggests that WNV

RNA replication and/or protein expression interferes with poly (I:C)-mediated IFN

gene induction in Hela cells (Scholle & Mason, 2005). Yet another report

indicates that WNV delays activation of interferon regulatory factor 3 (IRF-3) by

RIG-I early in infection to allow for virus replication (Fredericksen & Gale, 2006).

13

1.1.6 Diagnosis, treatment and prognosis

Virus isolation, detection of viral nucleic acid or antigen and seroconversion are

all recognized methods for diagnosis of recent WNV infection (Davis at a/.,

2006b), however, each method has its pros and cons. Due to biosafaty issues,

WNV isolation is not the first choice for WNV diagnosis. Viral nucleic acid

detection by polymerase chain reaction (PCR) amplification is useful for

screening blood products for potential contamination but not reliable enough for

diagnosis purposes due to the disappearance of viremia at the time disease

onset when the patient would submit blood samples (Davis at a/., 2006b).

Detection of WNV-specific antibodies in serum and CSF using commercially

available enzyme-linked immunosorbent assays (ELISA) are the methods of

choice for diagnosis of WNND (Tardei at a/., 2000). However, cross reaction

may result from previous exposure to other flaviviruses and/or recent vaccination

for yellow fever or Japanese encephalitis. Plaque reduction neutralization assay

will be necessary to exclude cross reaction and to confirm positive ELISA results

(Hayes & Gubler, 2006). Currently there is no guaranteed treatment for WNV

disease. However, multiple treatment regimes have shown promises and are

under clinical trials. Among which include treatments with IFN-a, corticosteroids,

and WNV-specific IgM derived from donor plasma with high levels of WNV

antibodies (Murray at al., 2006), however the efficacy and safety are undergoing

evaluation. Case report has documented successful treatment with IFN-a2b 3

weeks after WN meningoencephalitis onset in an 83-year old man (Lewis &

Amsden, 2007). Antisense nucleic acid compounds designed to inhibit WNV

14

replication have shown significant antiviral activity in vitro (Deas at a/., 2005;

Torrence at a/., 2006) and are now being tested in humans in clinical trials.

Potential options for therapy are continuously being discovered- high-throughput

screening of compounds for antiviral activity has identified triaryl pyrazoline as an

inhibitor of flavivirus replication in cell culture (Goodell at a/., 2006; Puig­

Basagoiti at a/., 2006) and experiments in mice indicates TNF-a antagonist might

be worthy as a therapy to prevent WNV neuroinvasion (Wang at aI., 2004).

Average recovery time for mild form of WNF sufferers is one week post onset of

symptoms but prognosis for patients with severe WNND diseases is poor (Davis

at a/., 2006b; Hayes & Gubler, 2006). Case fatality rates of WNND range from 4-

15% (Green at a/., 2005; Gubler, 2007). Full recovery is to be expected in those

who have WN meningitis with no focal neurologic involvement (Hayes & Gubler,

2006); however, individuals with WN encephalitis or AFP may experience severe

and prolonged deficits, such as fatigue, weakness, aching, depression

parkinsonism and tremor in AFP patients are common and it could last from

weeks to months or remain for life (Hayes & Gubler, 2006).

1.1.7. Vaccines and disease prevention

As of today, there is no specific WNV vaccine licensed for humans use; however,

extensive efforts in WNV vaccine development have yielded significant progress.

Two chimeric virus (WNVNFV and WNVIDEN4) vaccines, one recombinant DNA

plasmid vaccine and one killed virus vaccine are under phase I or II clinical trial

(Table 1) for human use. The two chimeric vaccines encompass the

15

incorporation of WNV E and PrM DNA sequences into a 17 -D yellow fever or a

dengue serotype-4 virus backbone. Other underdevelopment vaccines include

DNA vaccines containing WNV antigen or Kunjun antigen expression, and

recombinant vaccine using measles virus as a vector for WNV antigens (Hayes &

Gubler, 2006). Another chimeric vaccine of fused bacterial f1agellin (STF2 Delta)

to the WNV envelope E-III domain has shown prominent innate and adaptive

immunogenicity in mice without adjuvant (McDonald et aI., 2007). For veterinary

use, five WNV vaccines have been licensed for horses and domestic geese

(Kramer et a/., 2008) (Table 1).

On the individual level, avoidance of mosquito bites by limiting outdoor exposure

is the best preventive measure for not contracting WNV (Gubler et a/., 2000).

Otherwise, insect repellent should be worn on clothing and skin, especially during

peak feeding hour from dusk to dawn. Using N, N-diethyl-m-toluamide (DEET)­

containing repellents are the most safe and effective (Fradin, 1998; Hayes &

Gubler, 2006). Alternatives include oil of lemon eucalyptus, soybean oil, and

picaridin for skin and permethrin for clothing (Fradin & Day, 2002). On the

community and state level, vector control surveillance using larvicide and

adulticide can be effective in reducing mosquito populations and human WNV

infection (Barnard & Xue, 2004; Fradin & Day, 2002). Additional preventive

measures should be taken in the screening of donated blood in WNV endemic

areas to prevent transmission through transfusion (Custer et a/., 2004).

16

Table 1. Vaccines licensed or in clinical trials

Ptodua: ruune ComptlIlyondlor Insdtute. . .. I· .. ·.vtu:cinetype Stllt\IJi

Innovator® Fort Dodge Animal Health Killed virus L Recombitek® Merial Recombinant canarypox L

virus PreveNile TM Intervet Chimeric virus L

(WNVIYFV)

NA Kimron Veterinal}' Killed virus L Institute/Crucell

NA CDCIFort Dodge Animal Recombinant DNA L Health plasmid

Chimeravax"IM-West Nile Acambis Cbimeric virus CT-Il (WNVIYFV)

VRC-WNVDNA01O-OO-VP NIAIDINlli Recombinant DNA CT-I plasmid

WNIDEN4-3'deIta30 NIAIDINlli Chimeric virus CT-I (WNVIDEN4)

NA Crucell Killed virus CT-I

AbbreYi.tiODS: NA, inlb"""tion not ... U.ble; L, licensed for Veterinary 11'ie; YFv. yellow fever virus; Gr-II, clinicol triaJ, phose ll; cr-I, clinicol triaJ, phase I; DEN4, dengue4vinrs. Sources of infonlbltion: httpIlwww.fortdodgellvestock.coml, httpI/www.merlaLcoml, htlplllwww.Jme\WtlllllLcomf,httpllwww.cruceII.cnmI, htlplllwww.cllniaJltrials.goYl.

( Kramer LD et al., Annu Rev Entomol, 2008, 53: 61-81).

1.2 Blood-brain barrier (BBB) and viral Infection

1.2.1 Introduction to BBB ·and transmIgration pathways through BBB

Multiple brain barriers exist presumably to protect the central nervous system

from the dynamic fluctuation of biochemical environment in the peripheral

systems and to thus to sustain homeostasis within the CNS. The brain is

biochemically segregated from the periphery by three major barriers: the blood-

brain barrier in the brain parenchyma, the blood-CSF barrier in the choroid

plexus-epithelial and the blood-subarachnoid epithelial interfaces. During the

discovery of barrier between the brain and blood, the impermeable properties

17

were profoundly observed in the vasculature within the cortex and cerebellum

compared to capillaries of heart and skeletal muscles (Reese & Kamovsky,

1967). Following the initial finding, extensive studies have been generated on

BBB characteristics since.

The BBB is a highly regulated interface which separates blood borne entities

from the CNS. Unlike peripheral vascular endothelial cells, the brain vascular

endothelial cells (BVEC) of the BBB are not fenestrated. They have a higher

mitochondrial volume fraction and higher electrical resistance but a low number

of vesicles; in addition, they have specialized transport systems (Irie & Tavassoli,

1991). Under physiologic conditions. the BBB maintains homeostasis by ensuing

adequate supply of necessary nutrients such as oxygen and glucose for brain

cells as well as low level of surveillancelreplacement immune cells from blood

(Petty & Lo, 2002). A complete neurovascular unit conferring the BBB property

consists of vascular endothelium, perivascular astrocytes, basement membrane,

and pericytes that are in physical proximity to the endothelium (Persidsky et a/ .•

2006). Among these various components, brain vascular endothelial cells

(BVEC) are in the frontline directly interacting with a large variety of blood

constituents and regulating their passage into the brain (Petty & Lo, 2002). A

functional polarity exists in BVEC between the luminal (or apical) membrane

where tight junctions are located and the abluminal membrane where the

adherens junctions lie just above the basement membrane (Petty & Lo, 2002).

Passage of molecules across the endothelial cells can occur through the cells

18

(transcellular) or between adjacent cells (paracellular) (Persidsky at a/., 2006).

Factors that govern solutes crossing the BBB depend on whether the solute is

lipid soluble or charged and size. Lipophilic compounds with a molecular size

less than 15 AO (-3.5kDa) (Rubas at a/., 1996) such as nicotine, ethanol and

heroin can readily cross the BBB by passive diffusion while charged molecules

such as sodium ions or proteins cross the BBB slowly or not at all (Oldendorf,

1976). Crossing of glucose and amino acids require specific transporters present

on endothelial cells (Vorbrodt at a/., 2001; Xiang at a/., 2003). Receptors systems

have been found for several molecules including insulin, low-density lipoprotein,

insulin-like growth factors and transferrin (Banks, 2004; Moos at al., 2000;

Persidsky at al., 2006). Other than solutes, pathogenic microbes and immune

cells also enter the CNS through the BBB via transcellular andlor paracellular

crossing. Barrier properties of the BBB is attributed primarily to the presence of

tight junctions between endothelial cells and the few number of pinocytotiC

vesicles in the BVEC compared to vascular endothelial cells in other organs such

as the heart and skeletal muscles (Reese & Kamovsky, 1967). While pinocytotic

vesicles are associated with the ferrying of small amounts of fluid and solutes

across the cell wall, tight junctions between BVEC cells form extremely tight

seals characterized by the presence of tight junction proteins which disable

paracelluar passage of molecules (Banks, 2004; Moos at al., 2000; Persidsky at

al., 2006; Petty & Lo, 2002). Regulation of brain vasculature function is not only

by the biochemical milieu in the blood stream, but also by neuron and glial cells

19

regulate based on the metabolic requirements and changes in microenvironment

in the brain (Petty & Lo, 2002).

1.2.2 Dysfunction of BBB in viral infections

1.2.2.1 Tight junction proteins (T JP) and BBB permeability

BBB endothelial tight junctions are composed of a complex and elaborate

combination of (1) transmembrane and (2) cytoplasmic proteins linked to an

actin-based cytoskeleton (Petty & Lo, 2002). Tight junction transmembrane

proteins include claud ins, occludin and junctional adhesion molecules (JAMs).

Cytoplasmic proteins consist of zonula occludens (ZO) -1, 2, 3 and cingulin. See

schematic for distribution and proposed interactions between T JP in Fig. 3.

(1) Transmembrane (Integral) proteins

Claud ins belong to a superfamily of transmembrane proteins of size between 20-

24 kDa with four transmembrane segments (Petty & Lo, 2002). They form

dimmers and bind homotypically to other claudin molecules on the adjacent cell

membrane to form the primary seal. They also bind to and localize another

transmembrane protein occludin to the cellular junction (Kubota et a/., 1999).

Claudins are believed to be the primary backbone of tight junction strands while

occudin serves to enhance tightness (Kubota at a/., 1999). At least 24 claudins

proteins have been identified in mouse and humans (Persidsky at aI., 2006; Petty

& Lo, 2002), but their expression patterns vary among tissues; claudin-1, -3, and

20

-5 have been detected in cerebral microvascular endothelium (Hawkins & Davis,

2005).

Occludin is a 65-kDa transmembrane protein with four transmembrane domains

with both the amino and carboxy terminus located intracellularly. The cytoplasmic

C-terminal domain provides occludin connection with the cytoskeleton via

accessory proteins, ZO-1 and ZO-2 (Persidsky at a/., 2006). It has multiple

phosphorylation sites and the phosphorylation regulates its interaction with the

cell membrane proteins and regulates barrier permeability (Hirase at aI., 2001;

Sakakibara at a/., 1997; Wachtel at a/., 1999). Occludin is highly expressed in

BMVEC (Hirase et a/., 2001; Vorbrodt at a/., 2001) and is suggested to contribute

to the electrical resistance across the BBB as that high levels of occludin has

been shown to ensure high electrical resistance of epithelial cell monolayers

(McCarthy at a/., 1996).

JAM-1, -2, and -3 are IgG superfamily proteins. JAM is a transmembrane protein

with a singular domain of 40 kDa size (Martin-Padura et a/., 1998). JAM proteins

are expressed on endothelial and epithelial cells and on the surfaces of

leukocytes, erythrocytes and platelets (Mandell & Parkos, 2005). JAM mediates

cell-to-cell adhesion by means of homophilic and heterophilic interactions. Short

cytoplasmic tails of JAMs have been reported to bind to cytoplasmic protein such

as ZO-1 (Ebnet et a/., 2000). Though the direct role and mechanism of JAM in

the tight junction has not been identified, JAM has been implicated to involved in

21

a wide range of physiologic functions, including barrier function, leukocyte

migration, platelet activation, angiogenesis, and reovirus binding (Mandell &

Parkos, 2005).

(2) Cytoplasmic accessory proteins

Zonula occludens (ZO)-1,-2, -3 proteins are important cytoplasmic tight junction

phosphoproteins of approximately 160 kDa to 220 kDa in size and form

submembranous plaque of tight junctions (persidsky at al., 2006). They belong to

the membrane-associated and guanylate kinase proteins (MAGUK) family

(Anderson at al., 1995). ZO proteins contain PDZ and SH3 domains which are

involved in bridging transmembrane proteins such as claudins and occludin to

cytoskeletal proteins actin (Fanning at al., 1998).

Cingulin is another cytoplasmic phosphoprotein of tight junctions. It is about 140

kDa to 160 kDa is size and has been localized on the cytoplasmic surface.

Binding partners of cingulin include ZO-1, -2, -3, myosin and AF-6 (Petty & Lo,

2002). It is suggested that cingulin functions as a scaffold protein to link cytolysis

plaque proteins, i.e. ZO-1, -2, and -3, to the cytoskeletal protein myosin

(Cordenonsi at al., 1999; D'Atri at al., 2002).

22

Adherens

Fig. 3. Proposed interactions of junctional complex proteins in the capillary endothelial cells of blood-brain barrier. At tight junctions, claud Ins have been proposed to function as the backbone by binding to claudins on adjacent cells and thus forming a seal. Claudlns also bind to occludln, which functions as a regulatory protein. Occludin's presence in the membrane correlates with increased electrical resistance across the membrane and decreased permeability. Occludin Is thought to be Involved also in the fence function of tight junctions. JAM Is Involved in cell-cell adhesion and contributes to permeability control. A series of cytoplasmic proteins Z0-1, ZO-2 and Z0-3 form the submembranous plaque of tight junctions. These proteins belong to the MAGUK family (membrane-assoclated guanylate kinase proteins) and are involved In the coupling of transmembrane proteins to the cytoskeleton actin. Several cytoplasmic accessory proteins have been Identified at tight junctions, including AF6, 7H6 and cingulin; see the text for their potential structural and functional roles. The adherens junctions are composed of transmembrane glycoprotelns of the cadherin super family, which are linked to the cytoskeleton via cytoplasmic anchor proteins,l3-catenln, y-<:etenln (plakoglobin) and p120ctn that belong to the Armadillo protein family. poeatenin and y0{;8tenin bind to a-<:etenin, which Is an actin-binding and actin-bundling molecule linking the adhesive cadherinlcatenin complex to the F actin-based cytoskeleton. [Petty MA and Lo EH, prog Neurobiol. 2002, 66 (5):311-23)

The expression and proper distribution of T JP are important and have direct

effects in conferring BBB integrity. Increased permeability is the major

impairment or form of BBB dysfunction in many viral infections, especially in

those which manifest acute neuroinflammation such as meningitis and

23

encephalitis (Afonso at a/., 2007; Annunziata, 2003; Luabeya at a/., 2000). HIV­

associated damentia (HAD) has been extensively studied and much is being

learned about this devastating disease. TNF-a has been demonstrated to

augment BBB permeability and ease HIV-1 traversal through the paracellular

pathway (Nottet et a/., 1996). Increased BBB permeability in HIV has been

suggested to be a result of degradation of T JP including ZO-1, -2, claudin-1, -5

and occludin (Andras at a/., 2003; Boven at a/., 2000; Dallasta at a/., 1999;

Kanmogne at a/., 2005; Nottet at a/., 1996). Similarly, decreased expression of

ZO-1 protein has been shown in extended length of cerebral microvessels in SIV­

infected rhesus macaques (Luabeya at a/., 2000). In addition, increased

permeability of endothelial monolayer model has been demonstrated in HCMV

infection. Actin stress fiber formation and decreased expression of occludin and

vascular endothelial cadherin were observed (Bentz at a/., 2006).

1.2.2.2 MatTix metalloprotelnases (MMP) and TJP

The basal lamina of BVEC is composed of typical extracellular matrix (ECM)

components collagens, laminin, fibronectin, entactin, tenascin, thrombospondin,

heparin sulfate proteoglycans, and chondroitin sulfate proteoglycans (del Zoppo

& Hallenbeck, 2000; Lyons & Jones, 2007; Rascher at a/., 2002). Interactions

between matrix proteins and endothelial T JP can result in the modification of tight

junction functions. Collagen type IV, fibronectin, and laminin can influence the

expression of endothelial T JP such as occludin (Savettieri at a/., 2000).

Disruption of the ECM is strongly associated with increased BBB permeability in

24

pathological conditions (Jian Liu & Rosenberg, 2005; Rascher et aI., 2002). In

the CNS, matrix metalloproteinases (MMP), a family of over 20 zinc-dependent

structurally related endopeptidases, have been shown to degrade components of

the basal lamina (Leppert et a/., 2001), leading to disruption of the BBB, and

contribute to the neuroinflammatory response in many neurological diseases

(Ichiyama et a/., 2007; Jian Liu & Rosenberg, 2005; Leppert et a/., 2001; Mandai

et a/., 2003; Rosenberg, 2002; Saadoun et a/., 2007). Pre-inflammatory

cytokines such as TNF-a/p, IL-1 and -2 have been demonstrated to modulate the

expression and regulation of MMP (Ben David et a/., 2008; Lockwood et a/.,

2008; Seguin et a/., 2008). Increase in MMP-1, -2 and -9 activities were

associated with the reduction in collagen IV content, decreased barrier integrity,

enhanced permeability, and monocyte migration across the BBB monolayer

model treated with ethanol or acetaldehyde (Haorah et a/., 2008). Further,

elevated MMP-2, -7, and -9 expressions have been associated with HAD,

influenza virus and HCV, T JP degradation and BBB disruption (Conant et a/.,

1999; Eugenin et a/., 2006b; Ichiyama et a/., 2007; Saadoun et a/ .. 2007).

1.2.2.3 Cell adhesIon molecules (CAM) and immune cell transmIgration

CAM are cell surface molecules present on all cells, especially endothelial cells.

They are generally involved in cell-te-cell and cell-to-matrix interactions (Dittmar

et a/., 2008; Lyons & Jones, 2007). Common families of CAM include integrins,

cadherins, selectins, immunoglobulin superfamily (lgSF) among others (Lyons &

Jones, 2007). In the context of endothelium, CAM are important mediator of

leukocytes adhesion and transmigration through the endothelium to injured tissue

25

sites during inflammation (Golias at a/., 2007; Man at a/., 2007; Rao at a/., 2007),

thay are also involved in cell signaling in important developmental processes

such as growth, proliferation, organization and cell migration (Lyons & Jones,

2007). On the other hand CAM have been shown to take part in pathologic

conditions such as neoplasia and metastasis (Araki at a/., 2001; Barthel at a/.,

2007; Dittmar at a/., 2008; Lyons & Jones, 2007; Niu at a/., 2007; Shirai at a/.,

2003; Wrtz, 2008). Resting endothelial cells generally do not interact with

circulating leukocytes because they lack cell surface adhesion (Tesfamariam &

DeFelice, 2007). In the presence of cytokines in response to stimuli such as

virus or bacteria, endothelial cells become activated and have enhanced

expression of CAM thus are able to bind and activate binding of leukocytes

(Johnston & Butcher, 2002).

The immunoglobulin superfamily (lgSF) encompasses a diverse collection of

molecules that share the basic structure of the immunoglobulins and include

'families' of CAM such as the intercellular adhesion molecule family (ICAM) and

vascular adhesion molecule family (Lyons & Jones, 2007). They are

predominantly expressed on lymphocytes and other white blood cells, and are

important in inflammation and immune-based reactions (Springer, 1990). One of

the intercellular adhesion molecules, ICAM-1, is expressed on junctional

epithelium and appears to facilitate the intra-epithelial accumulation of T­

lymphocytes or neutrophils (Crawford & Hopp, 1990). Several tumours express

ICAM-1, including breast, lung and colon carcinomas (Araki at a/., 2001; Dowlati

26

et a/., 2008; Gallicchio et a/., 2008; Shirai et a/., 2003; Skelding et a/., 2008;

Zheng et a/., 2006). The level of both tissue and serum ICAM-1 expression has

been correlated to the pattem of spread and potential for metastases (Barthel et

a/., 2007; Niu et a/., 2007; Wang et a/., 2007) and reduced expression of ICAM in

adenoid cystic carcinoma of the head and neck has been associated with a

better prognosis (Shirai et a/., 2003).

Selectins are a group of single-chain transmembrane proteins found on platelets

(P-selectin), leukocytes (L-selectin) and endothelial cells (E-selectin, P-selectin)

(Tu et a/., 1999). They are characterized by a lectin-binding domain, EGF-like

repeat elements, and further repeat elements based on complement-binding

proteins. This family of molecules mediates the attachment of leukocytes and

platelets to the endothelium of blood vessels, and is involved in their migration

during inflammation (Tu et a/., 1999). They bind to sialylated and fucosylated

carbohydrate ligands presented and mediate initial capture, tethering, and rolling

along endothelium (McEver, 2002). L-selectin is expressed on most circulating

leukocytes and mediates lymphocyte rolling along high endothelial venules of

secondary lymphoid organs during chronic inflammation and secondary capture

by adhering leukocytes (Sperandio et a/., 2003). P-selectin is stored in Weibel­

Palade bodies of endothelial cells and in intracellular a-granules of platelets and

quickly released to the plasma membrane upon endothelial cell activation

(McEver, 2002). P- and E-selectins are expressed in acute as well as in

chronically inflamed endothelium and serve as rolling molecules for monocytes,

27

neutrophils, effector T cells, B cells, and natural killer cells (McEver, 2002). P­

selectin binds PSG-1 that is expressed by all neutrophils, monocytes, and

lymphocytes (McEver & Cummings, 1997). E-selectin binds PSGL-1 (Katayama

at al., 2003), C044 (Katayama at al., 2005) , E-selectin Jigand-1 (ESL-1) (Hidalgo

at al., 2007; Steegmaier at al., 1995) on myeloid cells and CD43 on T-helper 1

lymphocytes (Shimizu at 81., 1991).

The integrins represent the largest known family of CAM and are all integral

membrane glycoproteins expressed in different combinations by all cells (Hynes,

1992). Integrins all consist of an A and a B subunit. Currently, 16 different A

subunits, 8 B subunits and 24 combinations have been identified (Van Waes &

Carey, 1992). The two subunits collaborate to bind molecules in the extracellular

matrix, the specificity of which is determined by the cell type and the subunit

combination. Interactions between integrins and the matrix trigger a spectrum of

signals (Hynes, 2002). These have profound effects on cell survival, proliferation

and motility by altering the structure and functional activity of the cytoskeleton

(Carter at 81., 1990; Lyons & Jones, 2007)

Following insults such as physical impacts or invasion of pathogens, blood

leukocytes extravasate through endothelium and basal lamina into the tissue

sites of injury. It is a complex and tightly regulated process of innate and

adaptive immune responses. Chemoattractant-induced transmigration of

immune cells initiates the activation of inflammatory cascade (Butcher, 1991;

28

Springer, 1994). The multistep process starts with loose binding of leukocytes to

endothelial cell surface via the interactions between specific cell surface

adhesion molecules and their ligands expressed on either endothelial cells or

leukocytes (Johnston & Butcher, 2002; Vestweber, 2007). First, P-selectins

mediates capture while E-selectin stabilizes rolling on endothelial cells bind to

their ligands present on circulating leukocytes to slow them down and activate

them (Vestweber, 2007). Upon activation by cytokines and chemokines, integrins

on leukocytes bind to members of the IgSF family of CAM such as ICAM-1 and

VCAM-1 and result in firm adhesion. Completion of leukocytes extravasation is

mediated by binding to transmembrane receptors such as PECAM-1 (CD99) or

JAM molecules (Petri & Bixel, 2006). See Fig. 4.

--

Junctional odhoslon receplora

VI _.Ia

Non·JuncUonal odkc3fon receptor

Fig. 4. Adhesion receptors involved in the diapedesis process. The junctional and the non junctional adhesion receptors at endothelial cell contacts are depicted between different cells for optical clarity. The nectin related PVR was classified as adherens junction protein because of its similarity to the nectins. The two receptor-type tyrosine phosphatases VE-PTP and RPTP are indeed able to affect the function of VE-cadherin; whether they are indeed involved in the leukocyte diapedesis process has not yet been shown. (Vestweber 0 , Immunol Rev. 2007, 218: 178-96)

29

Inflammatory cytokines and chemokines activate endothelial cells to up-regulate

soluble CAM (Tesfamariam & DeFelice, 2007). Endothelial cells are the target of

infection by several pathogens, including viruses and bacteria that have been

implicated in dysregulation of vessel wall function (Tesfamariam & DeFelice,

2007). Viral pathogens such as herpes simplex virus (HSV) (Kim at a/., 2000;

Vercellotti, 1990), cytomegalovirus (CMV) (van Dam-Mieras at a/., 1992), and a

number of respiratory viruses (Visseren et a/., 2000) infect the endothelium can

trigger procoagulant activity, thrombotic complications, and induce expression of

CAM. Soluble factors released from infected monocytes induce expressions of

adhesion molecules VCAM-1 and E-selectin in human vascular cell culture

(Carlos at a/., 1991) and sPECAM in HIV-encephalitic brain tissue (Eugenin at

a/., 2006a), which leads to increased macrophage and leukocytes infiltration. In

addition, up-regulation of MCP-1 (CCL2) expression enhanced transmigration in

brain endothelium model (Eugenin at a/., 2006b). Endothelial cells also directly

participate in immune reaction where they present antigens to activated T cells

by upregulating the expression of MHC-encoded molecules (Bagai at a/., 2005;

Pober at a/., 2001; Shen at a/., 1997) which interacts with CD4+ and CD8+ T

lymphocytes and mediate the recruitment and trafficking of inflammatory

leukocytes (pober at a/., 2001). Further, proinflammatory cytokines trigger

activation of neutrophils and injury to the endothelium, perturbing endothelial

membrane morphology, cell matrix organization and vascular permeability

(Argenbright & Barton, 1992; Bratt & Palmblad, 1997).

30

Taken together, activation of brain endothelium triggered by infectious agents

typically result in the production and release of proinflammatory mediators by

blood cells as well as astrocytes, microglia and neurons. These immune

mediators induce expressions of CAM on endothelial cells and leukocytes,

leading to enhanced infiltration of blood immune cells into the brain.

Transmigration of immune cells can compromise barrier integrity and it can be

employed as a potential means of pathogen entry if the infiltrating immune cells

are infected, the "Trojan horse" entry mechanism, as suggested in neuroinvasion

of HIV-1 infection.

31

Chapter 2. Thesis Scope

2.1 Background for Research Question: Whether HBMVE cells playa role

in WNV-CNS entry?

WNV infection of the CNS tissues often results in severe neurological

complications if not death. WNV antigens are observed in the CNS tissue of

WNV encephalitic individuals by histological analysis. However, it is unclear how

WNV enters the CNS to infect brain cells. Leaky BBB has been demonstrated in

WNV infected-mice and is associated with increased viral load in the brain.

Further, high WNV viremia is correlated to early brain entry in immunodeficient

mice. Together, there is suggestive evidence that WNV, in part, can be spread

by the hematogenous route and potentially through the BBB with increased

permeability.

2.2 Objectives and Hypothesis

The objective of the proposed study is to characterize cellular and molecular

modulation of tight junction integrity induced by WNV infection of HBMVE cells,

which would influence subsequent transmigration of WNV into the CNS.

The central hypothesis is that WNV can infect HBMVE cells and modulate the

expressions of T JP and CAM, thereby leading to enhanced migration of virus

across the BBB.

32

2.3 Specific Alms

2.3.1 Specific Aim 1: To infect HBMVE cells with WNV NY99 and to determine

WNV replication kinetics. Infection with WNV in HBMVE cells will be examined

using immunofluorescence staining (lFS). WNV replication kinetics will be

analyzed using plaque assay and real-time reverse-transcriptase polymerase

chain reaction (qRT-PCR).

2.3.2 Specific Aim 2: To analyze the differential expressions of T JP, CAM and

MMP in HBMVE cells infected with WNV. Differential expressions of mRNA

transcripts of TJP, CAM and MMP will be assayed by qRT-PCR. Protein levels

of certain T JP and CAM will be analyzed using Western blotting and IFS.

2.4 Significance

CNS involvement of WNV infection is correlated to poor prognosis if not fatal,

however, there is no vaccine or specific antiviral treatment for WNV infection in

humans to date. Therefore it is important to delineate the pathways and

mechanisms underlying WNV-CNS entry in order to devise therapeutics to target

prevention or progression ofWNV spread.

This study will allow us to understand more specifically the role of HBMVE cells

in WNV dissemination and provide clues and insights about the potential route(s)

and the mechanism(s) by which WNV enters the brain.

33

Chapter 3. Methods

3.1 Experimental Design for Specific Aim 1: To Infect HBMVE cells with

WNV NY99 and to determine WNV replication kinetics.

3.1.1 Infection of HBMVE cells In 6-well plates and on cover slips

HBMVE cells (see 3.3.1) were seeded on coverslips in 24-well plates (6x104

cellslwell for immunofluorescence staining), or in 6-well plates (6x105 cellslwell

for viral kinetics and host gene expressions) or in T-25 tissue culture flask (8x105

cellslflask for protein extraction) 24 h prior to infection. At 80-90% confluency,

cells were either mock-infected with medium only, or with infectious WNV (see

3.3.2) at MOl 1, or 5. Infection with UV-inactivated virus was performed at MOl

5. During infection, excess medium in each weillflask was removed leaving

minimal volume to cover cells. Virus inoculums prepared at appropriate

concentrations in 100 III volume were than added into designated wellslflasks.

Well-plateslflasks were then retumed to incubator (37°C with 5% C02) for 1 h

adsorption. Each weillflask was then washed twice with 1X PBS to remove

unadsorbed virus followed by replenishment of fresh medium. Well plates and

flasks were then kept at 37DC with 5% CO2 until time of supematant collection, or

cell harvest at specific time points.

3.1.2 Collection of culture supernatants, harvesting of cells and fixation

of cells on cover slips

Non-cumulative and cumulative culture supematants were collected in

independent experiments at 0 (after second wash after infection), 6 and 12 h,

34

and at 01 through 5 after infection. At each collection, complete culture

supernatant was transferred from the well to a collection tube followed by

centrifugation at 14,000 rpm for 2 min to remove cell debris. Cell-free

supematant was then transferred to a new cryotube and stored at -SooC. Non­

cumulative culture supematant was collected from the same culture wells at

different time points. At each collection time point, the well was replenished with

2 mL of fresh medium. For cumulative culture supematant collection, complete

culture supematant was collected from individual well which was assigned for

only one specific time point, followed by cell harvesting at that particular time

point.

Cells for RNA extraction were harvested at 6 and 12 h, and at 01 through 4 after

infection. Cells in 6-well-plates were washed twice with 1X PBS and lysed

directly with 350 )JL of RL T lysis buffer solution from the Qiagen RNeasy Mini Kit

(Qiagen Cat# 74106) supplemented with 1% I3-mercaptoethanol and then

collected in cryotubes and stored at -SooC until further processing.

Cells for protein extraction were harvested at 12 h and at 01 through 4 after

infection. Cells in T-25 tissue culture flasks were washed twice with cold 1X PBS

and lysed with 130 )JL of Pierce lysis buffer (Pierce Cat# 78501) containing 1 %

protease inhibitor. After 5 min of incubation with lysis buffer at 4°C, cell lysate

was collected and centrifuged at 12,000 rpm (or 14,000 g) for 15 min before

supematant was transferred into a new collection tube and stored at -800 C until

further proceSSing.

35

Cells for IFS were fixed at 12 h, and at D1 through 5 after infection. Cells on

cover slips were fixed in 4% paraformaldehyde (PFA) diluted in 1x PBS for 10

min at RT and stored in 1x PBS at 4°C until use.

3.1.3 Detection of WNV on cover slips by Immunofluorescence

At days 1, 2 and 3 after infection, mock-infected control and WNV-infected (MOl

5) HBMVE cells on coverslips were fixed in 4% PFA solution for 10 min at RT,

washed twice in 1XPBS and were permeabilized in 0.5% TritonX 100 for 15 min.

The cover slips were blocked with 4% BSA in 1X PBS for 1 h, washed three

times in 0.1% BSA in 1X PBS and incubated first with monoclonal human anti­

WNVenv antibody (1 :800, a gift from CDC, Fort Collins) at 4°C ovemight and

then with Alexa Fluor 488 conjugated goat anti-mouse secondary antibody

(1:1000, Invitrogen Cat#A-11001). Post washing with 0.1% BSA in 1X PBS, the

cell nuclei were counterstained with bisbenzidine (1 ng/ml) before mounting onto

a slide with Vectashield mounting medium (Vector laboratories, Burlingame,

CA). Fluorescent cells were examined using a Zeiss Confocal Pascal equipped

with a Zeiss Axiovert 200 microscope, equipped with appropriate fluorescence

filters and objectives.

3.1.4 Quantitation of virus titer in supernatants by plaque assay

Non-cumulative cell-free supematants ofWNV-infected HBMVE cells at MOl 1 or

5 collected at 0, 6 and 12 h, and at D1 through 5 were used for viral titer

determination. Supematants from infected cells were serially diluted 0 to 10.12

times by 10-fold dilutions into 100 IJl volume with CSC-medium. Each 100 IJl of

36

the diluted supematant was then added onto a 100% confluent monolayer of

Vero cells grown in 6-well plates cultured in approximately 1 ml of M-199

medium. After addition of the diluted supematants, 6-well plates were gently

swirled to allow homogenous mixing of the supematant with the M-199 medium.

After mixing, Vero cells in 6-well plates were placed in the incubator for one hour

of incubation at 37°C and 5% C02. After one hour, Vero cells were overlaid with

3 ml of M-199 medium containing 1% agarose with gentle swirling to allow

mixing of agarose and the supematant before retuming to the incubator. Three

days later, each well was again overlaid with 3 ml of M-199 medium containing

1 % agarose and 1 % neutral red and retumed to the incubator. On the following

day, plaques were enumerated from each well. The dilution corresponding to the

lowest plaque count (below ten plaques) was the final viral titer expressed in

PFU/mL

3.1.5 Quantitation of virus RNA copy In supernatant by qRT·PCR

Cumulative cell-free supematant of WNV-infected HBMVE cells at MOl 1 or 5

collected at 0, 6 and 12 h, and at 01 through 5 were used for viral transcripts

analysis. Viral RNA from WNV-infected (MOl 1 and 5) cells was extracted (see

3.1.5.1) and reverse-transcribed into cDNA (see 3.1.5.2). cDNA was diluted

three times and 2 III was used as template for qRT-PCR assay for WNV

envelope gene RNA copy with probe which was expressed as PFUlmL Assay

standards templates were generated from RNA extracted from infectious WNV of

3x107 PFUI ml and reverse-transcribed into cDNA. Serial 1 a-fold dilutions of this

cDNA were then prepared to construct a standard curve ranges from 105 to 10.2

37

PFU for WNV RNA copy quantitation based on a fluorescent probe specific for

the WNV envelope gene. PCR thermal cycling was initiated with a first

denaturation step of 4 min at 95°C followed by 40 cycles of 95°C for 30s, 55°C

for 60 s and 68°C for 3 min. Each sample was assayed in duplicate for at least

two times.

3.1.5.1 extraction of viral RNA from supernatants

60 IJL of viral RNA was extracted from 140 IJL of cumulative and non-cumulative

cell-free culture supematant collected at 0,6 and 12 h, and at D1 through 5 using

QlAamp Viral RNA Mini Kit (Qiagen Cat # 52906) according to the instructions

provided by manufacturer.

3.1.5.2 Synthesis of cDNA from viral RNA from supernatants

15 IJL of viral RNA extracted was reverse-transcribed into cDNA using iScript

cDNA synthesis kit (Bio-Rad Cat# 170-8890) following the instructions provided

by the manufacturer. In brief, reaction mix, reverse transcriptase and RNA

template were mixed to a final reaction volume of 20 IJL by adding nuclease-free

water and were incubated for 40 min according to the reaction conditions

provided by the manufacturer.

3.1.6 Quant/tat/on of virus RNA copy in infected HBMVE cells by qRT-PCR

Viral RNA from the cell Iysates of WNV-infected (MOl 1 and 5) and UV­

inactivated WNV-infected cells (MOl 5) were extracted and reverse-transcribed

into cDNA as described previously (see 3.1.6.1 and 3.1.6.2). cDNA was diluted

38

three times and 2 III was used as template for qRT-PCR assay for WNV N54B

gene. A standard curve for the quantification of WNV RNA copy was constructed

using serial 10-fold dilutions of a plasmid containing the entire linear N54B

genome ranging from 108 to 103 copies. PCR thermal cycling was the same as

3.1.5. Each sample was assayed in duplicate for at least two times. In addition,

the same templates were also assayed by qRT-PCR wHh primer set specific for

WNV envelope gene and detection by fluorescence probe (see 3.1.5).

3.1.6.1 Extraction of cellular RNA from cell Iysates

At 6 and 12 h, and at D1 through 4 after infection, cells in 6-well plates were

washed twice wHh 1X PBS and lysed directly wHh 350 III of Rl T lysis buffer

solution supplemented wHh ~mercaptoethanol from the Qiagen RNeasy Mini Kit

(Qiagen Cat# 74106). 60 III of total cellular RNA was extracted from Iysates

using RNAeasy kit following manufacturer's spin protocol (Qiagen). Genomic

DNA contamination was minimized by digesting RNA wHh RNase-free DNase

twice before elution (Qiagen Cat#79254). RNA concentration and purity was

measured wHh a Spectrophotometer (Beckman Coulter DU 800, Fullerton, CA).

3.1.6.2 Synthesis of cDNA from cellular RNA

One microgram of cDNA was synthesized from 1 Ilg of cellular RNA using iScript

cDNA synthesis kit (Bio-Rad Cat# 170-8890) following the instructions provided

by the manufacturer. In brief, reaction mix, reverse transcriptase and RNA

template were mixed to a final reaction volume of 20 III by adding nuclease-free

39

water and were incubated for 40 min according to the reaction conditions

provided by the manufacturer.

3.2 Experimental Design for Specific Aim 2: To analyze the differential

expressions of CAM, T JP and MMP in HBMVE cells infected with

WNV.

3.2.1 Infection of HBMVE cells in 6-well plates, T-25 tissue culture flasks

and on cover slips

Same as 3.1.1

3.2.2 Harvesting of cells and fixation of cells on cover slips

Same as 3.1.2

3.2.3 Evaluation of transcription fold-change of genes of Interest by rea/­

tlmeRT-PCR

Fold-change in the expressions of T JP, CAM and MMP transcripts were

analyzed by real-time qRT-PCR. Cellular RNA from mock-infected, WNV­

infected (MOl 1 and 5) and UV-inactivated WNV-infected (MOl 5) cells were

extracted and reverse-transcribed into cDNA as described previously (see

3.1.6.1 and 3.1.6.2). cDNA was diluted three times and 2-4 IJL was used as

template for real-time PCR assay with genes of interest (ZO-1, claudin-1,

claudin-5, occludin, ICAM-1, VCAM-1, E-selectin, PECAM, MMP-2, -3, -9, and

TMIP-1). Two IJL of cDNA template from each sample was used for PCR with

GAPDH primer, house-keeping gene, to which our expression fold-change was

40

normalized. Relative expression in terms of fold-change compared to control

was determined using the Gene Expression Analysis Software accompanied with

iCycler iQ Real Time PCR Detection system (Bio-Rad). PCR thermal cycling

condition was the same as 3.1.5. Each sample was assayed in duplicate for at

least two times.

3.2.4 MMP activity assay

MMP-9 activity was assayed with the matrix metalloproteinase-9 (MMP-9)

Biotrak, Activity Assay System by Amersham Biosciences (GE Healthcare Cat#

RPN2634). MMP is captured by specific antibodies precoated onto a microplate.

Any active MMP activates the pro-detection enzyme, enabling it to cleave a

chromogenic peptide substrate. The resultant color is read at 405 nm and the

concentration of active MMP is extrapolated from a standard curve as detailed in

the instruction manual. One hundred IJL per cell culture supematant sample from

mock-infected and WNV Mal 1 and Mal 5-infected cells collected at 0,6, 12 h

and at D1 through 5 after infection was assayed following the protocol provided

by the assay system.

3.2.5 Detection of the expression of proteins of Interest by western blot

Total cellular protein was extracted from mock-infected and WNV-infected (Mal 1

and 5) cells. Cells in T-25 tissue culture flasks were washed twice with cold 1X

PBS and lysed with 130 IJL of Pierce lysis buffer (Pierce Cat# 78501) containing

1% protease inhibitor. After 5 min of incubation with lysis buffer at 4°C, cell

lysate was collected and centrifuged at 12,000 rpm (or 14,000 g) for 15 min

41

before supernatant was transferred into a new collection tube. Protein

concentration was assayed using the Quick Start Bradford Protein Assay (Bic­

Rad Cal# 500-0203) and 200 ~l of total protein was heat stabilized with reducing

agent at 95°C for 5 min. Equal amount of total protein (57 to 76 ng) from WNV­

infected cells and its paired mock-infected control were loaded and fractionated

on NuPAGE 4-12% Bis-Tris gel (Invitrogen Cal# NP0322Box) by electrophoresis

at 145 volts for 1-2 h depending on protein size. The gel was then transferred by

electro-blotting onto nitrocellulose membrane with 0.2 ~m pore size (Invitrogen

Cal# lC2000) or PVDF membrane with 0.45 ~m pore size (Invitrogen Cal#

lC2005) depending on the size of the protein of interest and blocked for 2 h in

5% BSA in 1x TBS with 0.05% Tween 20 (blocking solution). After six times of

washing in 1X TBST (wash buffer) on a shaker for 30 min, the membrane was

incubated with primary antibody for 2 h at room temperature in the blocking

solution followed by washing in wash buffer six times on a shaker for 30 min.

The membrane was then incubated with the appropriate secondary antibody in

the blocking solution for 1 h, followed by six washes for 30 min in the wash buffer

and two times in ddH20 before color developing using AP-conjugated substrate

kit (Bio-Rad Cal# 170-6432). Alternatively, enhanced chemiluminescence (ECl)

detection by ECl plus western blotting detection reagents (GE Healthcare Cal#

RPN2132) was used for protein bands development on Amersham Hyperfilm

(Amersham Cal# 28-9068-35) following provider's instructions.

42

3.3 Materials

3.3.1 HBMVE cells

HBMVE cells were purchased from Applied Cell Biology Research Institute

(ACBRI Cat# 376) (Kirkland, WA) at passage 2 (P2). Cells were passaged from

P2 up to P8 according to ACBRI protocol to maximize cell quantity to be used for

experiments. Cells were cultured in CS-C complete medium kit containing 10%

serum (Cell Systems, Cat# 4Z0-500) supplemented with 1 % gentamycin

sulphate solution (Cell Systems Cat# 4Z0-11 0). Seeding of cells in culture flasks

or well-plates was preceded by coating with attachment factor (Cell Systems

Cat# 4Z0-210) and passage procedure was performed following the instruction

that came with the Passage Reagent Group (Cell Systems Cat# 4Z0-800)

provided by Cell Systems Corporation. All experiments performed on HBMVE

cells were between 8 t010 passages, at which cells should retain their endothelial

cell properties according to company's recommendation. Except during

experimental procedures, cells were kept at 37°C with 5% C02 at all time.

3.3.2 WN virus

West Nile virus strain NY 99 used in this study was kindly provided by Dr. Duane

Gubler from the Department of Tropical Medicine, Medical Microbiology and

Pharmacology at the University of Hawaii, Manoa, Hawai'i, U.S.A. The virus was

originally isolated in 1999 from patients with West Nile encephalitis in New York,

subsequently propagated through horses, suckling mice and Vero cells. Stock

virus of 3x107 PFUlmL was diluted to proper concentrations for infection in all

43

experiments. For infection with UV-inactivated WNV, infectious WNV was diluted

in 500 III PBS in a 35-mm culture plate and exposed to UV radiation using a UV­

Strata linker 2400 device (Stratagene) for 10 min before infecting HBMVE cells.

3.3.3 Antibodies

For WNV antigen detection, monoclonal mouse anti-WNVlKunjun envelope

antibody was provided by Division of Vector-borne Infectious Diseases, CDC,

Fort Collins, Colorado. For tight junction protein IFS and Westem blotting, the

primary antibodies employed include: monoclonal mouse anti-ZO-1 (Zymed Cat#

33-9100), polycloncal rabbit anti-claudin-1 (Zymed Cat# 51-9000), monoclonal

human anti-VCAM-1 (R&D Cat# BBA5) for IFS, and polyclonal rabbit anti-VCAM-

1 (Santa Cruz Biotech Cat# 8304) for WB. Secondary antibodies employed

include: AlexaFluor 546 goat anti-mouse IgG (Invitrogen Cat# A 11003) for anti

WNV, Alexa Fluor 488 goat anti-mouse IgG (Invitrogen Cat# A-11001) for ZO-1

and VCAM-1, and Fluorescein-linked whole antibody from donkey anti-rabbit IgG

(Amersham Biosciences Cat# N1034) for claudin-1.

44

Chapter 4. Results

4.1 WNV infected and replicated in HBMVE cells

Whether WNV can infect and replicate in brain endothelium is unclear and this

information is pivotal in the elucidation of hematogenous spread of WNV via the

BBB route during high viremia. Therefore, we first examined the susceptibility of

HBMVE cells to WNV infection and the kinetics of virus replication upon infection.

Mock-infected and MOl S-infected HBMVE cells were fixed in 4% PFA at 12 h,

and at 01 through 4 after infection for WNV antigen IFS analysis. We observed

positive WNV antigen staining at all time points on coverslips. Fig SB

demonstrates positive WNV antigen staining (red) at 02 after infection which is

absent in mock-infected cells at the same time point (Fig. SA). Total numbers of

cells infected with WNV from 01 to 04 were estimated by counting

immunofluorescent cells positive for WNV from three coverslips for each time

point. Listed in Table 2 is the approximation of percentage of WNV-infected cells

obtained from counting 2,SOO to 3,SOO cells per coverslip totaled from S random

fields per coverslip. Numbers were average from three independent cover slips

from three independent experiments. Number of WNV-infected cells increased

from 3% at 01 to 4.S% at 02, where infection was at peak, and declined on 03

until it was almost undetectable at D4 after infection (Table 2).

45

Fig. 5 WNV could infect HBMVE cells. Mock-infected and WNV-infected (MOl 5) HBMVE cells on coverslips were stained with mouse anti-WNVenv primary antibody (1 :800) followed by rabbit anti-mouse Alexa Fluor 546 secondary antibody (1: 1 000) and counterstained with bisbenzidine for nuclear visualization (blue). (A) Mock-infected HBMVE cells do not show red fluorescence. (B) Fluorescence micrograph demonstrates infection of HBMVE cells with WNV evident by positive staining for WNVenv (red) at 02 after infection. (C) Magnified view of infected cells indicated by arrowhead in (B). Micrographs are representatives of three independent experiments performed in duplicate. Scale bar represents 20 IJm in A and B.

Table 2. Percent of WNV-infected cells by IFS staining

Oay after infection % infected cells Total cells counted

01 2.99 9579

02 4.55 8021

03 2.63 7709

04 0.24 7457

46

Replication kinetics of WNV in HBMVE cells was analyzed by profiling WNV titer

in the culture supernatants of infected cells collected at 0,6 and 12 h, and at 01

through 5 after infection. Infectious WNV titer in non-cumulative supernatant was

quantitated by both plaque assay as well as qRT-PCR. Plaque assay data

demonstrate robust increase in virion production from 6 h to 02 after infection at

both MOl 1 and MOl 5 infections. A steady 2-log10 PFU/mL incremental increase

in virus titer was observed from 6 h to 02 after infection at MOl 5 (Fig. 6). Peak

titers of 4x108 PFUlmL and 5x106 PFUlmL were detected for MOl 5 and MOl 1,

respectively, on 02 after infection. In addition, a 2-10910 PFU/mL difference

between the two infection doses is apparent only from 12 h up to 02 after

infection. Virus release declined drastically after peak release at 02 and reached

basal level of approximately 9x103 PFUlmL by 05 after infection (Fig. 6).

47

-~ 10 :; IL 11. -.. ! ;I

~ 3= 10

• MOl 10~------------~--~--------~----~~

Oh 6h 12h 01 02 03 04 05 Time after infection

Fig. 6 WNV titer recovered from cell culture supernatant quantitated by plaque assay. Non-cumulative cell-free culture supernatants collected from WNV-infected cells at O. 6 and 12 h, and at 01 through 4 after infection were diluted 0 to 10-12 folds and added to monolayer of Vero cells for viral titer determination based on visible plaque formation. Virus release increased rapidly starting from 6 h and peaked at 02 after· infection. Since 02 after infection, virus titer declined drastically and reached basal level by 05 after infection. Oata are representative of three independent experiments in duplicate. Error bars show SO.

48

Standard protocol adopted from the Division of Vector-Bome Infectious Diseases

of Centers for Disease Control and Prevention was employed for WNV RNA copy

determination by qRT-PCR. Viral RNA copy in PFU/mL based on WNV envelope

gene amplicon was quantitated in the cumulative culture supematants collected

from DO (0 h) through D4 after infection. These viral RNA transcripts represent

the amount of virus released up to the particular collection time point after

infection. Therefore, a profile of continuous increase is displayed reflecting the

accumulation of virus throughout the collection period (Fig. 7). Virus RNA copies

increased rapidly from DO until 02 after infection. Virus release dramatically

decreased after 02 after infection; however, a minute increase of less than 1-

IOg10 was detected on 03 throughout D4 after infection. Maximal virus release at

02 after infection is consistent with our plaque assay data (Fig. 6) as well as our

intracellular WNV RNA copy data (Fig. 8).

49

- 106

..J • MOl 1 E 106 -:::I • MOIS &L-a. 104 -fI) CD 'a 103 0 U

cr: 102 z a::: > 101 Z 3:

10° 00 01 02 03 04

Time after infection

Fig. 7 WNV transcripts recovered from cell culture supematant quantitated by qRT -peR. Viral RNA extracted from cumulative cell-free supematant collected from WNV-infected cells at DO (0 h) through 4 after infection was reverse-transcribed into cONA and amplified for virus RNA copy determination. Amplification of virus was conducted with primers and fluorescence probe specific for WNV envelope gene. Cumulative viral RNA copies increased rapidly from DO to 2 after infection and plateaued thereafter, although a minute increase of less than 1-logl0 was detected on 03 throughout D4 after infection. Data are representative of two independent experiments in duplicate. Error bars show SO.

50

After detection and quantitation of WNV in culture supernatants, infection of

HBMVE cells was also confirmed by intracellular quantitation of WNV NS4B (Fig.

BA) and envelope (Fig. BB) RNA transcripts. Cellular RNA from WNV-infected

cells (MOl 1 and MOl 5) and UV-inactivated WNV-infected cells (MOl 5) were

extracted and reverse-transcribed into cONA and amplified by qRT-PCR for WNV

NS4B and envelope gene expressions analysis. WNV RNA transcripts were

detected in all infected cells. However, only cells infected by WNV, but not by

UV-inactivated WNV, demonstrate intracellular virus replication (Fig. BA and B).

NS4B transcripts from MOl 5-infected cells increased most rapidly from 4x107

copies/llg RNA at 12 h to 1.5x109 copies/llg RNA at 01 after infection (Fig. BA)

and envelope transcripts increased from 9x103 PFUI IIg RNA at 12 h to 2x105

PFUI IIg RNA at 01 after infection (Fig. BB). A 1-logl0 decrease was detected

from 02 to 04 after infection (Fig. BA and B). As expected, WNV replication was

not detected in HBMVE cells infected with W-inactivated WNV where WNV RNA

transcripts continually decreased from 6 h to 04 after infection (Fig. BA and B).

51

1010~A------------------------------------------~

.... _-.- . ~- .--~- ~- ... . . .. . . .. . . . . . .. . . . .. .. .. . ..... ..

o 106~ ________________________________________ --,

B

~ ~ 104 --.--,,;:---===.-:3 It - e .. ..... - "e- .. . .

- --.-" -~-~-- ---. -. -- --_ .•. . --.... - ..... ~ MOl 1 - •• -- MOl 5 - - -e- •. UV MOl 5

D4 o +---------------------------------------~

6h 12h 01 02 03

TIme after Infection

Fig. 8 Intracellular WNV RNA transcripts quantitated by qRT -peR. One jJg of RNA extracted from the Iysates of infected cells harvested at 6 and 12 h, and at 01 through 4 after infection was reverse-transcribed into cDNA and two jJL of 1:3 diluted cDNA template of each sample was used for virus RNA copy quantitation based on a standard curve constructed by serial 10-fold dilutions of a plasmid containing the linear NS4B gene (A) or of the WNV envelope gene with a specific fluorescence probe (8). Consistent between the two protocols, WNV transcripts increased most rapidly from 12 h to 01 after infection. Maximal WNV transcripts were detected from 01 through 02 after infection and decreased thereafter. WNV replication was not detected in HBMVE cells infected with UV-

52

inactivated WNV. Data are representative of four independent experiments in duplicate. Error bars show SO.

53

4.2 Replication-competent WNV, but not W-Inact/vated WNV, differentially

up-regulated T JP expression In HBMVE cells.

Disruption of the BBB often involves T JP reduction or disorganization in the

event of pathogenic infection by viruses. Therefore, we investigated the effect on

TJP expressions in HBMVE cells infected with WNV. HBMVE cells were mock­

infected, infected with WNV at MOl 1 or MOl 5, or with UV-inactivated WNV at

MOl 5. Cellular RNA was extracted from cells harvested at 6, 12 h, and at D1

through 4 after infection, cDNA was synthesized and qRT-PCR was conducted to

analyze the expression of T JP, which was expressed as fold-change. All fold­

change was determined in comparison to mocked-infected control with GAPDH

being the house keeping gene for expression normalization. Four T JP: claudin-1

and -5, zonula occludens-1 (ZO-1) and occludin, were analyzed. Significant up­

regulation was detected only in claudin-1 expression but not in claudin-5 (Fig.

BA), ZO-1 or occludin (Fig. BB). Claudin-1 fold increase was significant in both

MOl 1 and MOl 5 infected cells at D3 and 4 after infection (Fig. BA) Maximal

increase in claudin-1 expression of 12-fold was detected in MOl 5-infected cells

at D3 after infection and it decreased to 8-fold at D4 after infection. Cells

infected with UV-inactivated WNV at MOl 5 did not yield any change in claudin-1

expression (Fig. BA), suggesting that the up-regulation observed was a direct

effect of WNV replication. Further, claudin-1 protein level of MOl 5-infected cells

was further examined by westem blot analysis (Fig. BC) and IFS (Fig. BE-F). As

substantiating support of our qRT-PCR data, claudin-1 protein levels were

significantly increased at D3 and 4 after infection compared to the corresponding

54

mock-infected control at each time point. Densitometric analysis of three

experiments confirmed an average of two to four-fold increase in claudin-1

expression on D3 and D4 respectively (Fig. 9D). IFS of claudin also revealed

increased immunoreactivity in WNV-infected HBMVE cells at D3 after infection

(Fig. 9F) compared to mock-infected control (Fig. 9E).

55

Q) C> C co ..c u

I "'0 o

u..

6h 12h 01 02 03 04

Time after infection

o CI-1 MOl 1 • CI-1 MOl 5 • CI-1 UV-MOI 5 !Ell CI-5 MOl 1 ~ CI-5 MOl 5

5 .---------------------------------------------~ B

4

3

2

1

o 6h 12h 01 02 03 04

Time after infection

El ZO-1 MOl1 I!!! ZO-1 MOl 5 0 Ocln MOl1 • Ocln MOl5

56

c 12 h

C I 01

C I 02

C I 03

C I 04

C I

Claduin-1 1 r--22 kDa ~~~~~~~~~~

p-actin 1_ - ,------ - - r42kDa

D

e 800 +' c: r

8 600

~ 400 ~ III ~ 200 0 c:

'#. 0

T T T

r 1 ) ;

12h D1 D2 D3 D4 Time after infection

57

Fig. 9 WNV differentially modulated T JP expressions quantitated by qRT­

peR, WB and IFS. cDNA template prepared as described in the materials and

methods was used for T JP expression expressed as fold-change compared to

mock-infected control at each time point normalizing to GAPDH expression

levels. (A) Claudin-1 was significantly up-regulated at both MOl 1 and 5 on D3

and D4 after infection. UV-inactivated WNV did not induce claudin-1 expression

in HBMVE cells. WNV did not induce changes in the mRNA expressions of

claudin-5 (A), ZO-1 or occuldin (8). Data are representative offour independent

experiments in duplicate. Error bars show SD. (C) Protein level of claudin-1 in

MOl 5-infected cells was analyzed by westem blot. Claudin-1 protein level

significantly increased at D3 and 4 after infection compared to the corresponding

mock-infected controls. j3-actin protein level demonstrated consistent loading

within each time point. (D) Densitometric analysis of claudin-1 from three

experiments. Western blot image is representative of three independent

experiments. IFS of Claudin-1 showed increased immunoreactivity in WNV­

infected HBMVE cells at D3 after infection (F) compared to mock-infected cells

(E).

58

4.3 Replication-competent WHit, but not UV-inactivated WHit, selectively

induced CAM expressions In HBMVE cells.

Cellular and molecular induction of CAM expression in HBMVE cells as a result

of WNV infection was investigated. Specifically, the expressions of VCAM-1,

ICAIYI-1, PECAM and E-selectin, which are commonly induced in other viral

infections, were analyzed. The same RNA derived-cDNA from cells harvested at

6, 12 h, and at D1 through 4 after infection were used for CAM expression fold­

change analysis using qRT-PCR. Fold-change was determined in comparison to

mocked-infected control with GAPDH being the house keeping gene for

expression normalization. Illustrated in Fig. 10A are the fold-change of VCAM-1

and E-selectin, which were significantly up-regulated at D2 to 4 after infection.

Maximal VCAM-1 expression increase of 3D-fold was detected at D2 after

infection in MOl 5 infected cells whereas maximal E-selectin fold increase of 10-

fold was detected at D3 after infection in both MOl 1- and MOl 5-infected cells.

Similar to claudin-1 up-regulation (Fig. 9A), induction of both VCAM-1 and E­

selectin expression was not detected in cells infected with UV-inactivated WNV

(Fig. 10A), confirming that the up-regulations observed were induced by WNV

replication. ICAM-1 and PECAM expressions were also analyzed, however, no

significant fold-change was detected (data not shown). Further, protein levels of

VCAM-1 in MOl 5-infected cells were further examined by western blot analysis

(Fig. 10B) and IFS (Fig 10D-E). Congruent with and lending support to our qRT­

PCR data, VCAM-1 protein levels were significantly increased from D2 to 4 after

infection compared to the corresponding mock-intected control at each time

59

point. Densitometric analysis of three experiments confirmed an average of four­

fold increase VCAM-1 expression on 02 and 04, and a 6-fold increase on 03

after infection (Fig. 10C). Positive IFS against VCAM-1 was only observed in

WNV-infected HBMVE cells on 03 after infection (Fig. 100) but not in mock­

infected HBMVE cells (Fig. 10E)

60

B

VCAM-1

l3-actin

6h

12 h

C I

12h 01 02 03 04

Time after infection

!8lI VCAM-1 MOl IE VCAM-1 MOl • VCAM-1 UV-MOI

~ E-selectin MOl ~ E-selectin MOl

D 1

C I

D2

C I

D3

C

D4

C

- 110kDa ~~~==~~~~==~ ..... _-_ ... -..tJ- 42kDa

61

c "0 ~ 1200

81000

(/) 800 > (!) (/) !II ~ 400 ()

c: 200

'#. 12h 01 02 03 04

Time after infection

Fig. 10 WNV selectively induced CAM expression quantitated by qRT-PCR, WB and IFS. cDNA template prepared as described in Fig. 8 was also used for CAM expression fold-change determination compared to mock-infected control at each time point normalized to GAPDH expression levels (A). VCAM-1 and E­selectin mRNA expressions in WNV-infected cells were significantly up-regulated for over 5 folds from D2 to 4 after infection . Maximum increase in VCAM-1 (30 folds) and E-selectin (10 folds) expressions were detected on D3 after infection at MOl 5 infection (A). Infection with UV-inactivated WNV at MOl 5 did not induce significant expression of VCAM-1 or E-selectin (A). Data are representative of four independent experiments in duplicate. Error bars show SD. Protein level of VCAM-1 from MOl 5-infected cells was analyzed by western blot (8 ). Consistent with the qRT-PCR data (A). VCAM-1 protein levels significantly increased from D2 throughout D4 after infection compared to the corresponding mock-infected controls (8). ~-actin protein level shown demonstrates consistent loading within each time point. Densitometric analysis of VCAM-1 from three experiments (C). Western blot image shown is representative of three independent experiments. IFS of VCAM-1 (green)

62

revealed immunoreactivity in WNV-infected cells at 03 after infection (E) which was not observed in mock-infected HBVME cells of the same time point (0).

63

4.4 Infection of HBMVE cells with WNV did not result in MMP or TIMP

expressions

MMP and its inhibitor TIMP are important matrix modulators and have been

documented to play roles in T JP degradation and BBB disruption leading to

increased vascular permeability during inflammatory conditions (Samuel &

Diamond, 2005). Since endothelial cells under pathological conditions have been

shown to secrete MMP-2, -3, -7 and -9 induced by inflammatory cytokines

(A~ona at al., 2007a; Wang at al., 2004), we examined the expressions of MMP-

2, -3, -9 and TIMP-1 by qRT-PCR. The same RNA derived-cDNA from cells

harvested at 6, 12 h, and at 01 through 4 after infection described under T JP and

CAM expression analyses was used for MMP and TIMP expression fold-change

analysis using qRT-PCR. All fold-change was determined in comparison to

mocked-infected control with GAPDH being the house keeping gene for

expression normalization. In contrast to our expectation, qRT-PCR analyses of

MMP and TIMP did not yield any changes in MMP and TIMP expressions

compared to mock-infected control (data not shown). In fact, the basal levels of

MMP and TIMP were very low when compared to a positive control derived from

stimulated blood cells. ELISA for MMP-9 activity was performed on non­

cumulative and cumulative supematants collected from 0, 6, and 12 h, and at 01

through 4 after infection. No difference in MMP-9 activity was observed between

experimental infected-samples and mock-infected control samples (data not

shown).

64

Chapter 5. Discussion

WNND occur in approximately 1 % of the infected individuals. While

meningoencephalitis is acute and can be fatal if not treated promptly, recovery

from other forms of WNND is often lengthy and symptoms may persistent

through life. Therefore, it is of most value to prevent WNV from entering the

CNS; however, the mechanism by which WNV enters the brain is not well

understood. While WNV can exploit multiple mechanisms to enter the CNS,

recent studies in mice have implicated the likelihood of WNV transmigration via

the BBB through hematogenous spread (Samuel & Diamond, 2005; Wang et a/.,

2004). Similar to polio virus CNS-invasion, Samuel et al has most recently

demonstrated that WNV is able to spread through antero- and retro-grade

transport from peripheral nerve axon into the CNS in hamsters (Samuel et a/.,

2007). As the various pathways and mechanisms of WNV CNS-entry are being

uncovered, we aimed to further elucidate the role of the BBB by examining the

response of HBMVE cells, the key component of the BBB, to WNV infection. Our

in vitro data are the first report of direct cellular and molecular modulations

induced by WNV infection in HBMVE cells. We demonstrated that HBMVE cells

are susceptible to WNV infection and are able to support viral replication without

causing noticeable cytopathic effects and cell death during the duration of our

study - five days. Further, we report increased expression of a tight junction

protein, claudin-1, and up-regulations of cell adhesion molecules, E-selectin and

VCAM-1, induced directly by WNV infection and replication.

65

WNV replication kinetics varies among cell types and has never been

characterized in HBMVE cells. In neurons, WNV titers determined by plaque

assay in vitro reached as high as 1x107 PFUlmL at day 6 after infection and

remained high until day 14 after infection while WNV titers in astrocytes and

microglia were only around 1 x1 05 and 1 x1 03 PFUlmL, respectively, which

dramatically decreased by day 5 after infection (Cheeran et s/., 2005). Our data

reveal WNV titers in HBMVE cells to be relatively high, like that in neurons, but

the replication cycle lasted only for five days, similar to that of astrocytes and

microglia. Therefore, infection of HBMVE cells, or of the BBB, is transient,

similar to the transient nature of WNV viremia, and detection of which may easily

be missed during the course of WNV infection.

Experimental productive infection of brain endothelial cells have been

documented in many neurotropic viruses, such as simian immunodeficiency virus

(SIV) (Mankowski et s/., 1994; Strelow et s/., 2002; Strelow et s/., 1998), human

cytomegalovirus (HCMV) (Bentz et 81., 2006; Lathey et s/., 1990), measles virus

(MV) (Cosby & Brankin, 1995), and equine herpes virus-1 (EHV-1) (Hasebe et

s/., 2006), which suggest transcellular migration of cell-free virus from the blood

into the CNS. When progeny virions are released on the abluminal side of the

infected endothelial cells, cell-free virus can make its way through the basal

lamina and have direct contact to resident brain cells such as astrocytes and

microglia and eventually neurons. Our demonstration of productive infection of

WNV in HBMVE cells precisely supports the same CNS entry mechanism.

Further, our result is supported by the occasional demonstration of WNV antigen

66

staining in gray owls (Lopes at a/., 2007) and crows (Wunschmann et a/., 2004)

in endothelial cells. Infection of human brain endothelium by Japanese

encephalitis virus, another closely related f1avivirus to WNV, also suggests the

positive role of HBMVE cells in virus spread (German et a/., 2006)

Infection with whole virus or exposure to certain viral proteins can elicit cellular

and molecular changes in HBMVE cells, such as SIV (Luabeya at a/., 2000;

Maclean at a/., 2005), HSV (Kim at a/., 2000) and HIV (Andras at a/., 2003;

Boven at a/., 2000; Dallasta at a/., 1999; Kanmogne at a/., 2005; Kanmogne at

a/., 2007). We examined the expressions of various important tight junction

proteins and cell adhesion molecules which have been commonly found to be

modulated in a variety of viral infections in the context of increased BBB

permeability. The continuous presence of tight junctions along the entire length

of brain vasculature is crucial to BBB property, thus the fundamental integrity of

brain endothelial tight junctions, which is in part governed by the expression and

distribution of TJP, are also important in conferring the BBB property of brain

endothelium. Unlike other viruses, WNV induced increased expression of

claudin-1 tight junction protein in HBMEV cells, but no change in claudin-5, ZO-1

and occludin. Interestingly, this unique up-regulation of claudin-1 was temporally

correlated with maximal virus replication and release as seen in our plaque assay

and qRT-PCR data, indicating that this tight junction modulation was mediated by

replication of WNV, rather than by the mere presence of viral proteins; no

increase was observed in HBMVE cells infected with UV-inactivated WNV.

67

Typically, viral infections of brain endothelial cells are associated with BBB

disruption mediated partly by reduction and/or disorganization of T JP as

described in HIV-1 (Andras et al., 2003; Oallasta et al., 1999; Kanmogne et al.,

2005), SIV (Luabeya et al., 2000; Maclean et al., 2005), and HCMV (Bentz et al.,

2006). To our knowledge, up-regulation of TJP has never been reported in the

context of viral infections; yet has been reported in cancers such as human

primary colon carcinoma (Ohawan et al., 2005; Kinugasa et al., 2007), oral

squamous cell carcinoma cells (Oku et al., 2006), and HPV-associated cervical

cancer (Vazquez-Ortiz et al., 2005). It is therefore perplexing for us to observe

an increase in claudin-1 expression. Based on the limited functional studies of

claudin-1 in the literature, we speculate that in the absence of immune

modulation, HBMVE cell's primary defensive response to WNV infection is

enhanced tight junction integrity via claudin-1 increase, possibly to prevent

further paracellular influx of virus. This conjecture is supported by in vitro

demonstration of approximately four times higher transendothelial resistance in

claudin-1-expressing cells than wild-type MOCK cells (Inai et al., 1999).

Furthermore, over-expression of claudin-1 was found to be associated with

reduced paracellular flux, suggesting major barrier function of claudin-1 protein

(Inai et al., 1999). Similar protective role of endothelial cells was previously

suggested for HIV-1 infection where in a BBB monolayer model, HBMVE cells

prevented HIV-1 from crossing the monolayer form one side to another (Fiala et

al., 1997). To determine the precise consequence of WNV-induced claudin-1

68

increase in HBMVE cells, further examination with an in vitro BBB model deems

necessary.

In addition to cell-free WNV transcellular migration through the BBB, it is also

possible that WNV crosses in cell-associated manner, i.e., the Trojan horse

mechanism. Infection of blood monoctye-derived macrophages by WNV has

been previously demonstrated in vitro (Rios et a/., 2006) and infection with

various viruses including HIV-1 (Kanmogne et a/., 2007; Nottet, 1999; Nottet et

a/., 1996; Song et a/., 2007) and HCMV (Bentz et a/., 2006) are often associated

with increased infiltration of infected or un infected leukocytes which may

contribute to endothelial tight junction disruption.

We report selective inductions of E-selectin and VCAM-1 in HBMVE cells by

WNV and that the induction temporally correlated with maximal virus replication

and release, indicating that endothelial cell activation is mediated by WNV

replication. Together, E-selectin and VCAM-1 are involved in increased

basophils, eosinophils and neutrophils adhesion (Bevilacqua et a/., 1989;

Bochner et a/., 1991; Hakkert et a/., 1991) to endothelial cells as well as

monocytes, macrophages and lymphocytes transmigration (Bentz et a/., 2006;

Nottet et a/., 1996; Wong et a/., 1999) across endothelium. Increased

expressions of E-selectin and VCAM-1 in HBMVE cells are likely to promote

interaction between circulating leukocytes and HBMVE cells given an in vivo

system. Thus, it is of potential that WNV may transmigrate through the BBB by

69

infecting leukocytes. Further, increased infiltration of macrophages and

lymphocytes are cardinal features documented in WNV encephalitic brain

histological studies in human (Hayes at al., 2005b), however, the underlying

mechanism of these immune cells influx are unclear. Based on our finding, we

believe that the perivascular immune cells infiltration can be partly modulated by

the increased expression of E-selectin and VCAM-1.

Increased BBB permeability can result from compromised tight junction as well

as leaky basal lamina, both mediated by increased MMP activities. As

suggested elsewhere, HBMVE cells can secrete MMP-2, -3, -7 and -9 in the

presence of pro-inflammatory stimuli such as TNF-a and IL-1 (Hummel at al.,

2001; Leppert at al., 2001). We also analyzed for the expressions of MMP-2, -3,

and -9, as well as an inhibitor of MMP-9, TIMP-1. Various MMP have been found

to cause T JP degradation as well as decomposition of the extra cellular matrix,

leading to increased permeability of the endothelium (Hawkins at al., 2007;

Rosenberg & Yang, 2007; Yang at al., 2007). In contrast to our expectation

based on the literature of other neurotropic viruses such as HIV-1 (Conant at al.,

1999; Eugenin at al., 2006b; Lafrenie at al., 1997; Sporer at al., 2000), analysis

of MMP-2, -3, -9 and TIMP-1 expressions revealed no change in HBMVE cells

infected with WNV. However, our MMP data are consistent with our T JP result

where no reduction was observed. Together, our result indicates that induction

of MMP is not a direct response of HBMVE cells to WNV infection but possibly an

indirect response under pathological conditions when pro-inflammatory mediators

70

are present. Therefore, further in vivo examination of the contribution of MMP in

BBB permeability changes with respect to WNV infection is warranted. Based on

our cell culture system, we have successfully delineated the direct cellular and

molecular response of HBMVE cells to WNV infection without complication of

host immune influence.

Taken together, our results suggest that HBMVE cells can play a role in the

dissemination of WNV from blood-to-brain. Direct WNV infection and replication

of HBMVE cells support transcellular crossing of cell-free WNV virus. As a direct

result of WNV replication, HBMVE cells up-regulated claudin-1 expression,

possibly to enhance tight junction integrity in order to fend off further infiltration of

virus and to prevent cell-t~1I spread. Also, WNV-induced expressions of E­

selectin and VCAM-1 are likely to promote adhesion and diapedesis of circulating

leukocytes across the BBB, thereby supporting paracellular entry of WNV via

"Trojan horse". Our study has characterized, on the cellular and molecular

levels, tight junction integrity of HBMVE cells infected with WNV. However, our

findings warrant further investigation on the functional increase of claudin-1, E­

selectin and VCAM-1 expressions with in vitro BBB model. In conclusion, our

study suggests an active role of HBMVE cells of the BBB in the dissemination of

WNV from the blood into the brain.

71

References

(2002a). Intrauterine West Nile virus infection-New York, 2002. Mmwr 51, 1135-

1136.

(2002b). Laboratory-acquired West Nile virus infections-United States, 2002.

Mmwr51,1133-1135.

(2002c). Possible West Nile virus transmission to an infant through breast­

feeding-Michigan, 2002. Mmwr 51,877-878.

Afonso, P. V., Ozden, 5., Prevost, M. C., Schmitt, C., Seilhean, D., Weksler, B.,

Couraud, P.O., Gessain, A., Romero, I. A. & Ceccaldi, P. E. (2007).

Human blood-brain barrier disruption by retroviral-infected lymphocytes:

role of myosin light chain kinase in endothelial tight-junction

disorganization. J Immunol179, 2576-2583.

Aktas, 0., U.llrich, 0., Infante-Duarte, C., Nitsch, R. & Zipp, F. (2007). Neuronal

damage in brain inflammation. Archives of neurology 64, 185-189.

Alexopoulou, L., Holt, A. C., Medzhitov, R. & Flavell, R. A. (2001). Recognition of

double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3.

Nature 413, 732-738.

Anderson, J. M., Fanning, A. 5., Lapierre, L. & Van ltallie, C. M. (1995). Zonula

occludens (ZO)-1 and ZO-2: membrane-associated guanylate kinase

homologues (MAGuKs) of the tight junction. Biochemical Society

transactions 23, 470-475.

72

Andras, I. E., Pu, H., Deli, M. A., Nath, A., Hennig, B. & Toborek, M. (2003). HIV-

1 Tat protein alters tight junction protein expression and distribution in

cultured brain endothelial cells. Journal of neuroscience research 74,255-

265.

Annunziata, P. (2003). Blood-brain barrier changes during invasion of the central

nervous system by HIV-1. Old and new insights into the mechanism.

Journal of neurology 250,901-906.

Araki, T., Miki, C. & Kusunoki, M. (2001). Biological implications of circulating

soluble intercellular adhesion molecule-1 in colorectal cancer patients.

Scandinavian journal of gastroenterology 36, 399-404.

Argenbright, l. W. & Barton, R. W. (1992). Interactions of leukocyte integrins with

intercellular adhesion molecule 1 in the production of inflammatory

vascular injury in vivo. The Shwartzman reaction revisited. The Journal of

clinical investigation 89, 259-272.

Arjona, A., Foellmer, H. G., Town, T., Leng, l., McDonald, C., Wang, T., Wong,

S. J., Montgomery, R. R., Fikrig, E. & Bucala, R. (2007a). Abrogation of

macrophage migration inhibitory factor decreases West Nile virus lethality

by limiting viral neuroinvasion. The Journal of clinical investigation 117,

3059-3066.

Arjona, A., Ledizet, M., Anthony, K., Bonafe, N., Modis, Y., Town, T. & Fikrig, E.

(2007b). West Nile Virus Envelope Protein Inhibits dsRNA-lnduced Innate

Immune Responses. J Immunol179, 8403-8409.

73

Armah, H. B., Wang, G., Omalu, B. I., Tesh, R. B., Gyure, K. A, Chute, D. J.,

Smith, R. D., Dulai, P., Vinters, H. V., Kleinschmidt-DeMasters, B. K. &

Wiley, C. A (2007). Systemic distribution of West Nile virus infection:

postmortem immunohistochemical study of six cases. Brain pathology

(Zurich, Switzerland) 17, 354-362.

Arndt, U., Wennemuth, G., Barth, P., Nain, M., AI-Abed, Y., Meinhardt, A,

Gemsa, D. & Bacher, M. (2002). Release of macrophage migration

inhibitory factor and CXCL8/interleukin-8 from lung epithelial cells

rendered necrotic by influenza A virus infection. Journal of virology 76,

9298-9306.

Bacher, M., Eickmann, M., Schrader, J., Gemsa, D. & Heiske, A. (2002). Human

cytomegalovirus-mediated induction of MIF in fibroblasts. Virology 299,

32-37.

Bagai, R., Valujskikh, A., Canaday, D. H., Bailey, E., Lalli, P. N., Harding, C. V. &

Heeger, P. S. (2005). Mouse endothelial cells cross-present Iymphocyte­

derived antigen on class I MHC via a TAP1- and proteasome-dependent

pathway. J Immunol174, 7711-7715.

Banks, W. A. (2004). The source of cerebral insulin. European journal of

pharmacology 490,5-12.

Barnard, D. R. & Xue, R. D. (2004). Laboratory evaluation of mosquito repellents

against Aedes albopictus, Culex nigripalpus, and Ochierotatus triseriatus

(Diptera: Culicidae). Journal of medical entomology 41, 726-730.

74

Barthel, S. R., Gavino, J. D., Descheny, l. & Dimitroff, C. J. (2007). Targeting

selectins and selectin ligands in inflammation and cancer. Expert opinion

on therapeutic targets 11, 1473-1491.

Beasley, D. W., Davis, C. T., Whiteman, M., Granwehr, B., Kinney, R. M. &

Barrett, A. D. (2004). Molecular determinants of virulence of West Nile

virus in North America. Arch Virol Suppl, 35-41.

Ben David, D., Reznick, A. Z., Srouji, S. & Livne, E. (2008). Exposure to pro­

inflammatory cytokines upregulates MMP-9 synthesis by mesenchymal

stem cells-derived osteoprogenitors. Histochemistry and cell biology.

Bentz, G. l., Jarquin-Pardo, M., Chan, G., Smith, M. S., Sinzger, C. & Yurochko,

A. D. (2006). Human cytomegalovirus (HCMV) infection of endothelial

cells promotes naive monocyte extravasation and transfer of productive

virus to enhance hematogenous dissemination of HCMV. Journal of

virology 80, 11539-11555.

Bevilacqua, M. P., Stengelin, S., Gimbrone, M. A., Jr. & Seed, B. (1989).

Endothelial leukocyte adhesion molecule 1: an inducible receptor for

neutrophils related to complement regulatory proteins and lectins. Science

(New York, NY243,1160-1165.

Bochner, B. S., Luscinskas, F. W., Gimbrone, M. A., Jr., Newman, W.,

Sterbinsky, S. A., Derse-Anthony, C. P., Klunk, D. & Schleimer, R. P.

(1991). Adhesion of human basophils, eosinophils, and neutrophils to

interleukin 1-activated human vascular endothelial cells: contributions of

75

endothelial cell adhesion molecules. The Journal of experimental medicine

173, 1553-1557.

Bode, A. V., Sejvar, J. J., Pape, W. J., Campbell, G. L. & Martin, A. A. (2006).

West Nile virus disease: a descriptive study of 228 patients hospitalized in

a 4-county region of Colorado in 2003. Clin Infect Dis 42, 1234-1240.

Boven, L. A., Middel, J., Verhoef, J., De Groot, C. J. & Nottet, H. S. (2000).

Monocyte infiltration is highly associated with loss of the tight junction

protein zonula occludens in HIV-1-associated dementia. Neuropathology

and applied neurobiology 26, 356-360.

Brabers, N. A. & Nottet, H. S. (2006). Role of the pro-inflammatory cytokines

TNF-alpha and IL-1beta in HIV-associated dementia. Europeanjoumal of

clinical investigation 36, 447-458.

Bragin-Sanchez, D. & Chang, P. P. (2005). West Nile virus encephalitis infection

in a heart transplant recipient: a case report. J Heart Lung Transplant 24,

621-623.

Bratt, J. & Palmblad, J. (1997). Cytokine-induced neutrophil-mediated injury of

human endothelial cells. J Immunol159, 912-918.

Brien, J. D., Uhrlaub, J. L. & Nikolich-Zugich, J. (2007). Protective capacity and

epitope specificity of CD8(+) T cells responding to lethal West Nile virus

infection. European journal of immunology 37, 1855-1863.

Brinton, M. A. (2002). The molecular biology of West Nile Virus: a new invader of

the western hemisphere. Annual review of microbiology 56,371-402.

76

Butcher, E. C. (1991). Leukocyte-endothelial cell recognition: three (or more)

steps to specificity and diversity. Cell 67, 1033-1036.

Campbell, G. L., Marfin, A. A., Lanciotti, R. S. & Gubler, D. J. (2002). West Nile

virus. The Lancet infectious diseases 2,519-529.

Carlos, T., Kovach, N., Schwartz, B., Rosa, M., Newman, B., Wayner, E.,

Benjamin, C., Osbom, L., Lobb, R. & Harlan, J. (1991). Human monocytes

bind to two cytokine-induced adhesive ligands on cultured human

endothelial cells: endothelial-leukocyte adhesion molecule-1 and vascular

cell adhesion molecule-1. Blood 77, 2266-2271.

Carter, W. G., Kaur, P., Gil, S. G., Gahr, P. J. & Wayner, E. A. (1990). Distinct

functions for integrins alpha 3 beta 1 in focal adhesions and alpha 6 beta

4/bullous pemphigoid antigen in a new stable anchoring contact (SAC) of

keratinocytes: relation to hemidesmosomes. The Joumal of cell biology

111,3141-3154.

Chambers, T. J. & Diamond, M. S. (2003). Pathogenesis of flavivirus

encephalitis. Advances in virus research 60, 273-342.

Chan, C. K., Limstrom, S. A., Tarasewicz, D. G. & Lin, S. G. (2006). Ocular

features of west nile virus infection in North America: a study of 14 eyes.

Ophthalmology 113,1539-1546.

Cheeran, M. C., Hu, S., Sheng, W. S., Rashid, A., Peterson, P. K. & Lokensgard,

J. R. (2005). Differential responses of human brain cells to West Nile virus

infection. Joumal of neurovirology 11, 512-524.

77

Chu, J. J. & Ng, M. L. (2004a). Infectious entry of West Nile virus occurs through

a clathrin-mediated endocytic pathway. Journal of virology 78, 10543-

10555.

Chu, J. J. & Ng, M. L. (2004b). Interaction of West Nile virus with alpha v beta 3

integrin mediates virus entry into cells. The Journal of biological chemistry

279,54533-54541.

Chu, J. J., Rajamanonmani, R., Li, J., Bhuvanakantham, R., Lescar, J. & Ng, M.

L. (2005). Inhibition of West Nile virus entry by using a recombinant

domain III from the envelope glycoprotein. The Journal of general virology

86,405-412.

Chung, K. M., Thompson, B. S., Fremont, D. H. & Diamond, M. S. (2007).

Antibody recognition of cell surface-associated NS1 triggers Fc-gamma

receptor-mediated phagocytosis and clearance of West Nile Virus-infected

cells. Journal of virology 81, 9551-9555.

Conant, K., McArthur, J. C., Griffin, D. E., Sjulson, L., Wahl, L. M. & Irani, D. N.

(1999). Cerebrospinal fluid levels of MMP-2, 7, and 9 are elevated in

association with human immunodeficiency virus dementia. Annals of

neurology 46,391-398.

Cordenonsi, M., D'Atri, F., Hammar, E., Parry, D. A., Kendrick-Jones, J., Shore,

D. & Citi, S. (1999). Cingulin contains globular and coiled-coil domains

and interacts with ZO-1, ZO-2, ZO-3, and myosin. The Journal of cell

biology 147, 1569-1582.

78

Cosby, S. L. & Brankin, B. (1995). Measles virus infection of cerebral endothelial

cells and effect on their adhesive properties. Veterinary microbiology 44,

135-139.

Crawford, J. M. & Hopp, B. (1990). Junctional epithelium expresses the

intercellular adhesion molecule ICAM-1. Joumal of periodontal research

25, 254-256.

Cushing, M. M., Brat, D. J., Mosunjac, M. I., Hennigar, R. A., Jemigan, D. B.,

Lanciotti, R., Petersen, L. R., Goldsmith, C., Rollin, P. E., Shieh, W. J.,

Guamer, J. & Zaki, S. R. (2004). Fatal West Nile virus encephalitis in a

renal transplant recipient. American joumal of clinical pathology 121, 26-

31.

Custer, B., Tomasulo, P. A., Murphy, E. L., Caglioti, S., Harpool, D., McEvoy, P.

& Busch, M. P. (2004). Triggers for switching from minipool testing by

nucleic acid technology to individual-donation nucleic acid testing for West

Nile virus: analysis of 2003 data to inform 2004 decision making.

Transfusion 44, 1547-1554.

D'Atri, F., Nadalutti, F. & Citi, S. (2002). Evidence for a functional interaction

between cingulin and ZO-1 in cultured cells. The Joumal of biological

chemistry 277,27757-27764.

Dallasta, L. M., Pisarov, L. A., Esplen, J. E., Werley, J. V., Moses, A. V., Nelson,

J. A. & Achim, C. L. (1999). Blood-brain barrier tight junction disruption in

human immunodeficiency virus-1 encephalitis. The American joumal of

pathology 155. 1915-1927.

79

Davis, C. W., Nguyen, H. Y., Hanna, S. L., Sanchez, M. D., Doms, R W. &

Pierson, T. C. (2006a). West Nile virus discriminates between DC-SIGN

and DC-SIGNR for cellular attachment and infection. Journal of virology

80,1290-1301.

Davis, L. E., DeBiasi, R, Goade, D. E., Haaland, K. Y., Harrington, J. A., Hamar,

J. B., Pergam, S. A., King, M. K., DeMasters, B. K. & Tyler, K. L. (2006b).

West Nile virus neuroinvasive disease. Annals of neurology 60, 286-300.

Deas, T. S., Binduga-Gajewska, I., Tilgner, M., Ren, P., Stein, D. A., Moulton, H.

M., Iversen, P. L., Kauffman, E. B., Kramer, L. D. & Shi, P. Y. (2005).

Inhibition of flavivirus infections by antisense oligomers specifically

suppressing viral translation and RNA replication. Journal of virology 79,

4599-4609.

del Zoppo, G. J. & Hallenbeck, J. M. (2000). Advances in the vascular

pathophysiology of ischemic stroke. Thrombosis research 98, 73-81.

Dhawan, P., Singh, A. B., Deane, N. G., No, Y., Shiou, S. R, Schmidt, C., Neff,

J., Washington, M. K. & Beauchamp, R D. (2005). Claudin-1 regulates

cellular transformation and metastatic behavior in colon cancer. The

Journal of clinical investigation 115, 1765-1776.

Diamond, M. S., Shrestha, B., Mehlhop, E., Sitati, E. & Engle, M. (2003a). Innate

and adaptive immune responses determine protection against

disseminated infection by West Nile encephalitis virus. Viral immunology

16,259-278.

80

Diamond, M. S., Sitati, E. M., Friend, L. D., Higgs, S., Shrestha, B. & Engle, M.

(2003b). A critical role for induced IgM in the protection against West Nile

virus infection. The Joumal of experimental medicine 198, 1853-1862.

Dittmar, T., Heyder, C., Gloria-Maercker, E., Hatzmann, W. & Zanker, K. S.

(2008). Adhesion molecules and chemokines: the navigation system for

circulating tumor (stem) cells to metastasize in an organ-specific manner.

Clinical & experimental metastasis 25, 11-32.

Dowlati, A., Gray, R, Sandler, A. B., Schiller, J. H. & Johnson, D. H. (2008). Cell

Adhesion Molecules, Vascular Endothelial Growth Factor, and Basic

Fibroblast Growth Factor in Patients with Non-Small Cell Lung Cancer

Treated with Chemotherapy with or without Bevacizumab-an Eastem

Cooperative Oncology Group Study. Clin Cancer Res 14,1407-1412.

Ebnet, K., Schulz, C. U., Meyer Zu Brickwedde, M. K., Pendl, G. G. & Vestweber,

D. (2000). Junctional adhesion molecule interacts with the PDZ domain­

containing proteins AF-6 and ZO-1. The Joumal of biological chemistry

275, 27979-27988.

Engle, M. J. & Diamond, M. S. (2003). Antibody prophylaxis and therapy against

West Nile virus infection in wild-type and immunodeficient mice. Joumal of

virology 77, 12941-12949.

Eugenin, E. A., Gamss, R, Buckner, C., Buono, D., Klein, R S., Schoenbaum, E.

E., Calderon, T. M. & Berman, J. W. (2006a). Shedding of PECAM-1

during HIV infection: a potential role for soluble PECAM-1 in the

pathogenesis of NeuroAIDS. Joumal of leukocyte biology 79, 444452.

81

Eugenin, E. A., Osiecki, K., Lopez, L., Goldstein, H., Calderon, T. M. & Berman,

J. W. (2006b). CCL2Imonocyte chemoattractant protein-1 mediates

enhanced transmigration of human immunodeficiency virus (HIV)-infected

leukocytes across the blood-brain barrier: a potential mechanism of HIV­

CNS invasion and NeuroAIDS. J Neurosci 26,1098-1106.

Fanning, A. S., Jameson, B. J., Jesaitis, L. A. & Anderson, J. M. (1998). The tight

junction protein ZO-1 establishes a link between the transmembrane

protein occludin and the actin cytoskeleton. The Joumal of biological

chemistry 273,29745-29753.

Ferguson, D. D., Gershman, K., LeBailly, A. & Petersen, L. R. (2005).

Characteristics of the rash associated with West Nile virus fever. Clin

Infect Dis 41,1204-1207.

Fiala, M., Looney, D. J., Stins, M., Way, D. D., Zhang, L., Gan, X., Chiappelli, F.,

Schweitzer, E. S., Shapshak, P., Weinand, M., Graves, M. C., Witte, M. &

Kim, K. S. (1997). TNF-alpha opens a paracellular route for HIV-1 invasion

across the blood-brain barrier. Molecular medicine (Cambridge, Mass 3,

553-564.

Fradin, M. S. (1998). Mosquitoes and mosquito repellents: a clinician's guide.

Annals of intemal medicine 128, 931-940.

Fradin, M. S. & Day, J. F. (2002). Comparative efficacy of insect repellents

against mosquito bites. The New England joumal of medicine 347, 13-18.

Fredericksen, B. L. & Gale, M., Jr. (2006). West Nile virus evades activation of

interferon regulatory factor 3 through RIG-I-dependent and -independent

82

pathways without antagonizing host defense signaling. Journal of virology

80,2913-2923.

Fredericksen, B. L., Keller, B. C., Fornek, J., Katze, M. G. & Gale, M., Jr. (2008).

Establishment and maintenance of the innate antiviral response to West

Nile Virus involves both RIG-I and MDA5 signaling through IPS-1. Journal

of virology 82, 609-616.

Gallicchio, M., Rosa, A. C., Dianzani, C., Brucato, L., Benetti, E., Collino, M. &

Fantozzi, R. (2008). Celecoxib decreases expression of the adhesion

molecules ICAM-1 and VCAM-1 in a colon cancer cell line (HT29). British

journal of pharmacology 153, 870-878.

Garcia-Tapia, D., Hassett, D. E., Mitchell, W. J., Jr., Johnson, G. C. & Kleiboeker,

S. B. (2007). West Nile virus encephalitis: sequential histopathological and

immunological events in a murine model of infection. Journal of

neurovirology 13, 130-138.

German, A. C., Myint, K. S., Mai, N. T., Pomeroy, I., Phu, N. H., Tzartos, J.,

Winter, P., Collett, J., Farrar, J., Barrett, A., Kipar, A., Esiri, M. M. &

Solomon, T. (2006). A preliminary neuropathological study of Japanese

encephalitis in humans and a mouse model. Transactions of the Royal

Society of Tropical Medicine and Hygiene 100, 1135-1145.

Girard, Y. A., Popov, V., Wen, J., Han, V. & Higgs, S. (2005). Ultrastructural

study of West Nile virus pathogenesis in Culex pipiens quinquefasciatus

(Diptera: Culicidae). Journal of medical entomology 42, 429-444.

83

Glass, W. G., Lim, J. K., Cholera, R., Pletnev, A. G., Gao, J. L. & Murphy, P. M.

(2005). Chemokine receptor CCR5 promotes leukocyte trafficking to the

brain and survival in West Nile virus infection. The Journal of experimental

medicine 202,1087-1098.

Glass, W. G., McDermott, D. H., Lim, J. K., Lekhong, S., Yu, S. F., Frank, W. A.,

Pape, J., Cheshier, R. C. & Murphy, P. M. (2006). CCR5 deficiency

increases risk of symptomatic West Nile virus infection. The Journal of

experimental medicine 203, 35-40.

Golias, C., Tsoutsi, E., Matziridis, A., Makridis, P., Batistatou, A. &

Charalabopoulos, K. (2007). Review. Leukocyte and endothelial cell

adhesion molecules in inflammation focusing on inflammatory heart

disease. In vivo (Athens, Greece) 21, 757-769.

Goodell, J. R., Puig-Basagoiti, F., Forshey, B. M., Shi, P. Y. & Ferguson, D. M.

(2006). Identification of compounds with anti-West Nile Virus activity.

Journal of medicinal chemistry 49, 2127-2137.

Green, M. S., Weinberger, M., Ben-Ezer, J., Bin, H., Mendelson, E., Gandacu,

D., Kaufman, Z., Dichtiar, R., Sobel, A., Cohen, D. & Chowers, M. Y.

(2005). Long-term Death Rates, West Nile virus epidemic, Israel, 2000.

Emerging infectious diseases 11, 1754-1757.

Guarner, J., Shieh, W. J., Hunter, S., Paddock, C. D., Morken, T., Campbell, G.

L., Martin, A. A. & Zaki, S. R. (2004). Clinicopathologic study and

laboratory diagnosis of 23 cases with West Nile virus encephalomyelitis.

Human pathology 35, 983-990.

84

Gubler, D. J. (2007). The continuing spread of West Nile virus in the western

hemisphere. Clin Infect Dis 45, 1039-1046.

Gubler, D. J., Campbell. G. L., Nasci. R.. Komar, N., Petersen. L. & Roehrig, J. T.

(2000). West Nile virus in the United States: guidelines for detection,

prevention, and control. Viral immunology 13, 469-475.

Hakkert, B. C., Kuijpers, T. W., Leeuwenberg, J. F., van Mourik, J. A. & Roos, D.

(1991). Neutrophil and monocyte adherence to and migration across

monolayers of cytokine-activated endothelial cells: the contribution of

CD18, ELAM-1, and VLA-4. Blood 78, 2721-2726.

Haorah, J., Schall, K., Ramirez, S. H. & Persidsky, Y. (2008). Activation of

protein tyrosine kinases and matrix metalloproteinases causes blood-brain

barrier injury: Novel mechanism for neurodegeneration associated with

alcohol abuse. Glia 56, 78-88.

Hasebe, R., Kimura, T., Nakamura, K., Ochiai, K., Okazaki, K., Wada, R. &

Umemura, T. (2006). Differential susceptibility of equine and mouse brain

microvascular endothelial cells to equine herpesvirus 1 infection. Archives

of virology 151, 775-786.

Hawkins, B. T. & Davis, T. P. (2005). The blood-brain barrier/neurovascular unit

in health and disease. Pharmacological reviews 57,173-185.

Hawkins, B. T., Lundeen, T. F., Norwood, K. M., Brooks, H. L. & Egleton, R. D.

(2007). Increased blood-brain barrier permeability and altered tight

junctions in experimental diabetes in the rat contribution of

hyperglycaemia and matrix metalloproteinases. Diabetologia 50, 202-211.

85

Hayes, E. B. & Gubler, D. J. (2006). West Nile virus: epidemiology and clinical

features of an emerging epidemic in the United States. Annual review of

medicine 57,181-194.

Hayes, E. B., Komar, N., Nasci, R S., Montgomery, S. P., O'Leary, D. R &

Campbell, G. L. (2005a). Epidemiology and transmission dynamics of

West Nile virus disease. Emerging infectious diseases 11, 1167-1173.

Hayes, E. B., Sejvar, J. J., Zaki, S. R, Lanciotti, R S., Bode, A. V. & Campbell,

G. L. (2005b). Virology, pathology, and clinical manifestations of West Nile

virus disease. Emerging infectious diseases 11, 1174-1179.

Hidalgo, A., Peired, A. J., Wild, M. K., Vestweber, D. & Frenette, P. S. (2007).

Complete identification of E-selectin ligands on neutrophils reveals distinct

functions of PSGL-1, ESL-1, and CD44. Immunity 26, 477-489.

Hirase, T., Kawashima, S., Wong, E. Y., Ueyama, T., Rikitake, Y., Tsukita, S.,

Yokoyama, M. & Staddon, J. M. (2001). Regulation of tight junction

permeability and occludin phosphorylation by Rhoa-p160ROCK­

dependent and -independent mechanisms. The Journal of'biological

chemistry 276, 10423-10431.

Hummel, V., Kallmann, B. A., Wagner, S., Fuller, T .• Bayas. A.. Tonn. J. C .•

Benveniste, E. N., Toyka, K. V. & Rieckmann, P. (2001). Production of

MMPs in human cerebral endothelial cells and their role in shedding

adhesion molecules. Journal of neuropathology and experimental

neurology 60, 320-327.

86

Hynes, R. O. (1992). Integrins: versatility, modulation, and signaling in cell

adhesion. Cell 69, 11-25.

Hynes, R. O. (2002). Integrins: bidirectional, allosteric signaling machines. Cell

110,673-687.

Ichiyama, T., Morishima, T., Kajimoto, M., Matsushige, T., Matsubara, T. &

Furukawa, S. (2007). Matrix metalloproteinase-9 and tissue inhibitors of

metalloproteinases 1 in influenza-associated encephalopathy. The

Pediatric infectious disease joumal 26, 542-544.

Inai, T., Kobayashi, J. & Shibata, Y. (1999). Claudin-1 contributes to the epithelial

barrier function in MOCK cells. European jot,lmal of cell biology 78, 849-

855.

lrie, S. & Tavassoli, M. (1991). Transendothelial transport of macromolecules: the

concept of tissue-blood barriers. Cell Bioi Rev 25.317-333.340-311.

Jean CM, H. S., Louie JK, Glaser CA. Risk factors for West Nile virus

neuroinvasive disease, Califomia, 2005. Emerging infectious diseases

[Epub; ahead of print].

Jian Liu, K. & Rosenberg, G. A. (2005). Matrix metalloproteinases and free

radicals in cerebral ischemia. Free radical biology & medicine 39, 71-80.

Johnston, B. & Butcher, E. C. (2002). Chemokines in rapid leukocyte adhesion

triggering and migration. Seminars in immunology 14, 83-92.

Johnston, L. J., Halliday, G. M. & King, N. J. (2000). Langerhans cells migrate to

local lymph nodes following cutaneous infection with an arbovirus. The

Joumal of investigative dermatology 114, 560-568.

87

Kanmogne, G. D., Primeaux, C. & Grammas, P. (2005). HIV-1 gp120 proteins

alter tight junction protein expression and brain endothelial cell

permeability: implications for the pathogenesis of HIV-associated

dementia. Journal of neuropathology and experimental neurology 64, 498-

505.

Kanmogne, G. D., Schall, K., Leibhart, J., Knipe, B., Gendelman, H. E. &

Persidsky, Y. (2007). HIV-1 gp120 compromises blood-brain barrier

integrity and enhances monocyte migration across blood-brain barrier:

implication for viral neuropathogenesis. J Cereb Blood Flow Metab 27,

123-134.

Katayama, Y., Hidalgo, A., Cha~g, J., Peired, A. & Frenette, P. S. (2005). CD44

is a physiological E-selectin ligand on neutrophils. The Journal of

experimental medicine 201, 1183-1189.

Katayama, Y., Hidalgo, A., Furie, B. C., Vestweber, D., Furie, B. & Frenette, P. S.

(2003). PSGL-1 participates in E-selectin-mediated progenitor homing to

bone marrow: evidence for cooperation between E-selectin ligands and

alpha4 integrin. Blood 102, 2060-2067.

Kelley, T. W., Prayson, R. A., Ruiz, A. I., lsada, C. M. & Gordon, S. M. (2003).

The neuropathology of West Nile virus meningoencephalitis. A report of

two cases and review of the literature. American journal of clinical

pathology 119, 749-753.

Kim, Y. C., Bang, D., Lee, S. & Lee, K. H. (2000). The effect of herpesvirus

infection on the expression of cell adhesion molecules on cultured human

88

dermal microvascular endothelial cells. Journal of dermatological science

24,38-47.

Kinugasa, T., Huo, Q., Higashi, D., Shibaguchi, H., Kuroki, M., Tanaka, T.,

Futami, K., Yamashita, Y., Hachimine, K., Maekawa, S., Nabeshima, K.,

Iwasaki, H. & Kuroki, M. (2007). Selective up-regulation of claudin-1 and

claudin-2 in colorectal cancer. Anticancer research 27, 3729-3734.

Kleiboeker, S. B., Loiacono, C. M., Rottinghaus, A., Pue, H. L. & Johnson, G. C.

(2004). Diagnosis of West Nile virus infection in horses. J Vet Diagn Invest

16,2-10.

Klein, R. S., Lin, E., Zhang, B., Luster, A. D., Tollett, J., Samuel, M. A., Engle, M.

& Diamond, M. S. (2005). Neuronal CXCL 10 directs CD8+ T-cell

recruitment and control of West Nile virus encephalitis. Joumal of virology

79, 11457-11466.

Kleinman, S., Glynn, S. A., Busch, M., Todd, D., Powell, L., Pietrelli, L., Nemo,

G., Schreiber, G., Bianco, C. & Katz, L. (2005). The 2003 West Nile virus

United States epidemic: the America's Blood Centers experience.

Transfusion 45, 469-479.

Kleinschmidt-DeMasters, B. K., Marder, B. A., Levi, M. E., Laird, S. P., McNutt, J.

T., Escott, E. J., Everson, G. T. & Tyler, K. L. (2004). Naturally acquired

West Nile virus encephalomyelitis in transplant recipients: clinical,

laboratory, diagnostic, and neuropathological features. Archives of

neurology 61, 1210-1220.

89

Kramer, L. D., Styer, L. M. & Ebel, G. D. (2008). A Global Perspective on the

Epidemiology of West Nile Virus. Annu Rev Entomol53, 61-81.

Kubota, K., Furuse, M., Sasaki, H., Sonoda, N., Fujita, K., Nagafuchi, A. &

Tsukita, S. (1999). Ca(2+)-independent cell-adhesion activity of claudins,

a family of integral membrane proteins localized at tight junctions. Curr

BioI 9, 1035-1038.

Kuno, G., Chang, G. J., Tsuchiya, K. R, Karabatsos, N. & Cropp, C. B. (1998).

Phylogeny of the genus Flavivirus. Joumal of virology 72, 73-83.

Lafrenie, R M., Wahl, L. M., Epstein, J. S., Yamada, K. M. & Dhawan, S. (1997).

Activation of monocytes by HIV-Tat treatment is mediated by cytokine

expression. J Immunol159, 4077-4083.

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. & Roehrig,

J. T. (2002). Complete genome sequences 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. & Gubler, D. J. (1999). Origin of the West Nile virus

responsible for an outbreak of encephalitis in the northeastem United

States. Science (New York, NY 286,2333-2337.

90

Lathey, J. L., Wiley, C. A., Verity, M. A. & Nelson, J. A. (1990). Cultured human

brain capillary endothelial cells are permissive for infection by human

cytomegalovirus. Virology 176, 266-273.

Lee, J. W., Chu, J. J. & Ng, M. L. (2006). Quantifying the specific binding

between West Nile virus envelope domain III protein and the cellular

receptor aiphaVbeta3 integrin. The Journal of biological chemistry 281,

1352-1360.

Leppert, D., Lindberg, R. L., Kappos, L. & Leib, S. L. (2001). Matrix

metalloproteinases: multifunctional effectors of inflammation in multiple

sclerosis and bacterial meningitis. Brain research 36, 249-257.

Lewis, M. & Amsden, J. R. (2007). Successful treatment of West Nile virus

infection after approximately 3 weeks into the disease course.

Pharmacotherapy 27, 455-458.

Lockwood, C. J., Oner, C., Uz, Y. H., Kayisli, U. A., Huang, S. J., Buchwalder, L.

F., Murk, W., Funai, E. F. & Schatz, F. (2008). Matrix Metalloproteinase 9

(MMP9) Expression in Preeclamptic Decidua and MMP9 Induction by

Tumor Necrosis Factor Alpha and Interleukin 1 Beta in Human First

Trimester Decidual Cells. Biology of reproduction.

Lopes, H., Redig, P., Glaser, A., Armien, A. & Wunschmann, A. (2007). Clinical

findings, lesions, and viral antigen distribution in great gray owls (Strix

nebulosa) and barred owls (Strix varia) with spontaneous West Nile virus

infection. Avian diseases 51,140-145.

91

Luabeya, M. K., Dallasta, L. M., Achim, C. L., Pauza, C. D. & Hamilton, R. L.

(2000). Blood-brain barrier disruption in simian immunodeficiency virus

encephalitis. Neuropathology and applied neurobiology 26, 454-462.

Lyons, A. J. & Jones, J. (2007). Cell adhesion molecules, the extracellular matrix

and oral squamous carcinoma. International journal of oral and

maxillofacial surgery 36,671-679.

Maclean, A. G., Belenchia, G. E., Bieniemy, D. N., Moroney-Rasmussen, T. A. &

Lackner, A. A. (2005). Simian immunodeficiency virus disrupts extended

lengths of the blood-brain barrier. Journal of medical primatology 34, 237-

242.

Man, S., Ubogu, E. E. & Ransohoff, R. M. (2007). Inflammatory cell migration into

the central nervous system: a few new twists on an old tale. Brain

pathology (Zurich, Switzerland) 17, 243-250.

Mandai, M., Mandai, A., Das, S., Chakraborti, T. & Sajal, C. (2003). Clinical

implications of matrix metalloproteinases. Molecular and cellular

biochemistry 252, 305-329.

Mandell, K. J. & Parkos, C. A. (2005). The JAM family of proteins. Advanced

drug delivery reviews 57,857-867.

Mankowski, J. L., Spelman, J. P., Ressetar, H. G., Strandberg, J. D., Laterra, J.,

Carter, D. L., Clements, J. E. & Zink, M. C. (1994). Neurovirulent simian

immunodeficiency virus replicates productively in endothelial cells of the

central nervous system in vivo and in vitro. Journal of virology 68, 8202-

8208.

92

Martin-Padura, I., Lostaglio, S., Schneemann, M., Williams, L., Romano, M.,

Fruscella, P., Panzeri, C., Stoppacciaro, A., Ruco, L., Villa, A., Simmons,

D. & Dejana. E. (1998). Junctional adhesion molecule. a novel member of

the immunoglobulin superfamily that distributes at intercellular junctions

and modulates monocyte transmigration. The Joumal of cell biology 142,

117-127.

McCarthy. K. M .• Skare. I. B .• Stankewich. M. C., Furuse, M., Tsukita, S., Rogers,

R. A .• Lynch, R. D. & Schneeberger, E. E. (1996). Occludin is a functional

component of the tight junction. Joumal of cell science 109 ( Pt 9), 2287-

2298.

McEver, R. P. (2002). Selectins: lectins that initiate cell adhesion under flow.

Current opinion in cell biology 14, 581-586.

McEver, R. P. & Cummings, R. D. (1997). Role of PSGL-1 binding to selectins in

leukocyte recruitment. The Joumal of clinical investigation 100, S97-103.

Mehlhop. E. & Diamond, M. S. (2008). The molecular basis of antibody protection

against West Nile virus. Current topics in microbiology and immunology

317, 125-153.

Mehlhop, E., Whitby, K., Oliphant, T., Marri, A., Engle, M. & Diamond, M. S.

(2005). Complement activation is required for induction of a protective

antibody response against West Nile virus infection. Joumal of virology 79,

7466-7477.

Moos, T., Trinder, D. & Morgan, E. H. (2000). Cellular distribution of ferric iron,

ferritin. transferrin and divalent metal transporter 1 (DMT1) in substantia

93

nigra and basal ganglia of normal and beta2-microglobulin deficient

mouse brain. Cellular and molecular biology (Noisy-Ie-Grand, France) 46,

549-561.

Mukhopadhyay, S., Kim, B. S., Chipman, P. R., Rossmann, M. G. & Kuhn, R. J.

(2003). Structure of West Nile virus. Science (New York, NY 302, 248.

Mukhopadhyay, S., Kuhn, R. J. & Rossmann, M. G. (2005). A structural

perspective of the flavivirus life cycle. Nature reviews 3, 13-22.

Murgue, B., Zeller, H. & Deubel, V. (2002). The ecology and epidemiology of

West Nile virus in Africa, Europe and Asia. Current topics in microbiology

and immunology 267,195-221.

Murray, K., Baraniuk, S., Resnick, M., Arafat, R., Kilborn, C., Cain, K.,

Shallenberger, R., York, T. L., Martinez, D., Hellums, J. S., Hellums, D.,

Malkoff, M., Elgawley, N., McNeely, W., Khuwaja, S. A. & Tesh. R. B.

(2006). Risk factors for encephalitis and death from West Nile virus

infection. Epidemiology and infection 134, 1325-1332.

Niu, J. X., Zhang, W. J., Ye, L. Y., Wu, L. Q., Zhu, G. J., Yang, Z. H., Grau, G. E.

& Lou, J. N. (2007). The role of adhesion molecules, alpha v beta 3, alpha

v beta 5 and their ligands in the tumor cell and endothelial cell adhesion.

Eur J Cancer Prev 16, 517-527.

Nottet, H. S. (1999). Interactions between macrophages and brain microvascular

endothelial cells: role in pathogenesis of HIV-1 infection and blood - brain

barrier function. Journal of neurovirology 5, 659-669.

94

Nottet, H. 5., Persidsky, Y., Sasseville, V. G., Nukuna, A. N., Bock. P., Zhai, Q.

H., Sharer, l. R., McComb, R. D., Swindells, 5., Soderland, C. &

Gendelman, H. E. (1996). Mechanisms for the transendothelial migration

of HIV-1-infected monocytes into brain. J Immunol156, 1284-1295.

Nybakken, G. E., Oliphant, T., Johnson,S., Burke,S., Diamond, M. S. &

Fremont, D. H. (2005). Structural basis of West Nile virus neutralization by

a therapeutic antibody. Nature 437,764-769.

Oku, N., Sasabe, E., Ueta, E., Yamamoto, T. & Osaki, T. (2006). Tight junction

protein claudin-1 enhances the invasive activity of oral squamous cell

carcinoma cells by promoting cleavage of laminin-5 gamma2 chain via

matrix metalloproteinase (MMP)-2 and membrane-type MMP-1. Cancer

research 66, 5251-5257.

Oldendorf, W. (1976). Blood-brain barrier. New York: Spectrum publications.

Pealer, l. N., Martin, A. A., Petersen, l. R., lanciotti, R. 5., Page, P. l., Stramer,

S. l., Stobierski, M. G., Signs, K., Newman, B., Kapoor, H., Goodman, J.

l. & Chamberland, M. E. (2003). Transmission of West Nile virus through

blood transfusion in the United States in 2002. The New England joumal

of medicine 349,1236-1245.

Persidsky, Y., Ramirez, S. H., Haorah, J. & Kanmogne, G. D. (2006). Blood-brain

barrier: structural components and function under physiologic and

pathologic conditions. J Neuroimmune Pharmacol 1, 223-236.

Petri, B. & Bixel, M. G. (2006). Molecular events during leukocyte diapedesis.

The FEBS joumal 273, 4399-4407.

95

Petty, M. A. & Lo, E. H. (2002). Junctional complexes of the blood-brain barrier:

permeability changes in neuroinflammation. Progress in neurobiology 68,

311-323.

Pober, J. 5., Kluger, M.S. & 5chechner, J. 5. (2001). Human endothelial cell

presentation of antigen and the homing of memory/effector T cells to skin.

Annals of the New York Academy of Sciences 941,12-25.

Puig-Basagoiti, F., Tilgner, M., Forshey, B. M., Philpott,S. M., Espina, N. G.,

Wentworth, D. E .• Goebel,S. J., Masters. P. 5., Falgout, B., Ren, P.,

Ferguson, D. M. & 5hi, P. Y. (2006). Triaryl pyrazoline compound inhibits

f1avivirus RNA replication. Antimicrobial agents and chemotherapy 50,

1320-1329.

Purtha, W. E., Myers, N., Mitaksov, V., 5itati, E., Connolly, J., Fremont, D. H.,

Hansen, T. H. & Diamond, M.S. (2007). Antigen-specific cytotoxic T

lymphocytes protect against lethal West Nile virus encephalitis. European

journal of immunology 37, 1845-1854.

Rao, R. M., Yang, L., Garcia-Cardena, G. & Luscinskas, F. W. (2007).

Endothelial-dependent mechanisms of leukocyte recruitment to the

vascular wall. Circulation research 101,234-247.

Rappole, J. H., Derrickson,S. R. & Hubalek, Z. (2000). Migratory birds and

spread of West Nile virus in the Western Hemisphere. Emerging infectious

diseases 6, 319-328.

Rascher, G., Fischmann, A., Kroger,S., Duffner, F., Grote, E. H. & Wolburg, H.

(2002). Extracellular matrix and the blood-brain barrier in glioblastoma

96

multiforme: spatial segregation of tenascin and agrin. Acta

neuropathologica 104, 85-91.

Reese, T. S. & Kamovsky, M. J. (1967). Fine structural localization of a blood­

brain barrier to exogenous peroxidase. The Journal of cell biology 34, 207-

217.

Rios, M., Zhang, M. J., Grinev, A., Srinivasan, K., Daniel, S., Wood, 0., Hewlett,

I. K. & Dayton, A. I. (2006). Monocytes-macrophages are a potential target

in human infection with West Nile virus through blood transfusion.

Transfusion 46, 659-667.

Rosenberg, G. A. (2002). Matrix metalloproteinases in neuroinflammation. Glia

39, 279-291.

Rosenberg, G. A. & Yang, Y. (2007). Vasogenic edema due to tight junction

disruption by matrix metalloproteinases in cerebral ischemia.

Neurosurgical focus 22, E4.

Rubas, W., Cromwell, M. E., Shahrokh, Z., Villagran, J., Nguyen, T. N., Wellton,

M., Nguyen, T. H. & Mrsny, R. J. (1996). Flux measurements across

Caco-2 monolayers may predict transport in human large intestinal tissue.

Journal of pharmaceutical sciences 85, 165-169.

Saadoun, D., Bieche, I., Authier, F. J., Laurendeau, I., Jambou, F., Piette, J. C.,

Vidaud, M., Maisonobe, T. & Cacoub, P. (2007). Role of matrix

metalloproteinases, proinflammatory cytokines, and oxidative stress­

derived molecules in hepatitis C virus-associated mixed cryoglobulinemia

vasculitis neuropathy. Arthritis and rheumatism 56, 1315-1324.

97

Sakakibara, A., Furuse, M., Saitou, M., Ando-Akatsuka, Y. & Tsukita, S. (1997).

Possible involvement of phosphorylation of occludin in tight junction

formation. The Journal of cell biology 137,1393-1401.

Samuel, M. A. & Diamond, M. S. (2005). Alphalbeta interferon protects against

lethal West Nile virus infection by restricting cellular tropism and

enhancing neuronal survival. Journal ofvirology 79,13350-13361.

Samuel, M. A. & Diamond, M. S. (2006). Pathogenesis of West Nile Virus

infection: a balance between virulence, innate and adaptive immunity, and

viral evasion. Journal of virology 80, 9349-9360.

Samuel, M. A., Wang, H., Siddharthan, v., Morrey, J. D. & Diamond, M. S.

(2007). Axonal transport mediates West Nile virus entry into the central

nervous system and induces acute flaccid paralysis. Proceedings of the

National Academy of Sciences of the United States of America 104,

17140-17145.

Savettieri, G., Di Liegro, I., Catania, C., Licata, L., Pitarresi, G. L., D'Agostino, S.,

Schiera, G., De Caro, V., Giandalia, G., Giannola, L. I. & Cestelli, A.

(2000). Neurons and ECM regulate occludin localization in brain

endothelial cells. Neuroreport 11,1081-1084.

Scholle, F. & Mason, P. W. (2005). West Nile virus replication interferes with both

poly(I:C)-induced interferon gene transcription and response to interferon

treatment. Virology 342,77-87.

98

Seguin, C. A., Pilliar, R. M., Madri, J. A. & Kandel, R. A. (2008). TNF-alpha

induces MMP2 gelatinase activity and MT1-MMP expression in an in vitro

model of nucleus pulposus tissue degeneration. Spine 33, 356-365.

Shen, J., SS, T. T., Schrieber, L. & King, N. J. (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. Joumal of virology 71, 9323-

9332.

Shimizu, Y., Shaw, S., Graber, N., Gopal, T. V., Horgan, K. J., Van Seventer, G.

A. & Newman, W. (1991). Activation-independent binding of human

memory T cells to adhesion molecule ELAM-1. Nature 349, 799-802.

Shirai, A., Furukawa, M. & Yoshizaki, T. (2003). Expression of intercellular

adhesion molecule (ICAM)-1 in adenoid cystic carcinoma of the head and

neck. The Laryngoscope 113,1955-1960.

Shrestha, B. & Diamond, M. S. (2004). Role of CD8+ T cells in control of West

Nile virus infection. Joumal of virology 78, 8312-8321.

Shrestha, B., Samuel, M. A. & Diamond, M. S. (2006). CD8+ T cells require

pertorin to clear West Nile virus from infected' neurons. Joumal of virology

80,119-129.

Sitati, E., McCandless, E. E., Klein, R. S. & Diamond, M. S. (2007). CD40-CD40

ligand interactions promote trafficking of CD8+ T cells into the brain and

protection against West Nile virus encephalitis. Joumal of virology 81,

9801-9811.

99

Sitati, E. M. & Diamond, M. S. (2006). CD4+ T-cell responses are required for

clearance of West Nile virus from the central nervous system. Journal of

virology 80, 12060-12069.

Skelding, K. A., Barry, R. D. & Shafren, D. R. (2008). SystemiC targeting of

metastatic human breast tumor xenografls by Coxsackievirus A21. Breast

cancer research and treatment.

Smithbum K, H. T., Burke A, Paul J (1940). A neurotropic virus isolated from the

blood of a native of Uganda. The American journal of tropical medicine

and hygiene 20,471-492.

Solomon, T. (2004). Flavivirus encephalitis. The New England journal of

medicine 351, 370-378.

Song, H. Y., Ryu, J., Ju, S. M., Park, L. J., Lee, J. A., Choi, S. Y. & Park, J.

(2007). Extracellular HIV-1 Tat enhances monocyte adhesion by up­

regulation of ICAM-1 and VCAM-1 gene expression via ROS-dependent

NF-kappaB activation in astrocytes. Experimental & molecular medicine

39,27-37.

Sperandio, M., Smith, M. L., Forlow, S. B., Olson, T. S., Xia, L., McEver, R. P. &

Ley, K. (2003). P-selectin glycoprotein Iigand-1 mediates L-selectin­

dependent leukocyte rolling in venules. The Journal of experimental

medicine 197,1355-1363.

Sporer, B., Koedel, U., Paul, R., Kohleisen, B., Erfle, V., Fontana, A. & Pfister, H.

W. (2000). Human immunodeficiency virus type-1 Nef protein induces

100

blood-brain barrier disruption in the rat: role of matrix metalloproteinase-9.

Journal of neuroimmunology 102,125-130.

Springer, T. A. (1990). Adhesion receptors ofthe immune system. Nature 346,

425-434.

Springer, T. A. (1994). Traffic signals for lymphocyte recirculation and leukocyte

emigration: the multistep paradigm. Cell 76, 301-314.

Steegmaier, M., Levinovitz, A., lsenmann, S., Borges, E., Lenter, M., Kocher, H.

P .• Kleuser. B. & Vestweber. D. (1995). The E-selectin-Iigand ESL-1 is a

variant of a receptor for fibroblast growth factor. Nature 373, 615-620.

Stramer, S. L., Fang, C. T .• Foster, G. A., Wagner, A. G., Brodsky, J. P. & Dodd,

R. Y. (2005). West Nile virus among blood donors in the United States,

2003 and 2004. The New England journal of medicine 353, 451-459.

Strelow, L., Janigro. D. & Nelson, J. A. (2002). Persistent SIV infection of a

blood-brain barrier moclel. Journal of neurovirology 8, 270-280.

Strelow, L. I., Watry, D. D., Fox, H. S. & Nelson, J. A. (1998). Efficient infection of

brain microvascular endothelial cells by an in vivo-selected neuroinvasive

SIVmac variant. Journal of neurovirology 4, 269-280.

Tardei. G .• Ruta. S .. Chitu, V., Rossi, C .• Tsai. T. F. & Cernescu. C. (2000).

Evaluation of immunoglobulin M (lgM) and IgG enzyme immunoassays in

serologic diagnosis of West Nile Virus infection. Journal of clinical

microbiology 38, 2232-2239.

Tesfamariam, B. & DeFelice, A. F. (2007). Endothelial injury in the initiation and

progression of vascular disorders. Vascular pharmacology 46. 229-237.

101

Tilleux, S. & Hermans, E. (2007). Neuroinflammation and regulation of glial

glutamate uptake in neurological disorders. Journal of neuroscience

research 85, 2059-2070.

Tilley, P. A., Fox, J. D., Jayaraman, G. C. & Preiksaitis, J. K. (2007).

Maculopapular rash and tremor are associated with West Nile fever and

neurological syndromes. Journal of neurology, neurosurgery, and

psychiatry 78, 529-531.

Torrence, P. F., Gupta, N., Whitney, C. & Morrey, J. D. (2006). Evaluation of

synthetic oligonucleotides as inhibitors of West Nile virus replication.

Antiviral research 70, 60-65.

Tu, L., Murphy, P. G., Li, X. & Tedder, T. F. (1999). L-selectin ligands expressed

by human leukocytes are HECA-452 antibody-clefined carbohydrate

epitopes preferentially displayed by P-selectin glycoprotein Iigand-1. J

ImmunoI163,5070-5078.

van Dam-Mieras, M. C., Muller, A. D., van Hinsbergh, V. W., Muliers, W. J.,

Bomans, P. H. & Bruggeman, C. A. (1992). The procoagulant response of

cytomegalovirus infected endothelial cells. Thrombosis and haemostasis

68, 364-370.

van der Meulen, K. M., Pensaert, M. B. & Nauwynck, H. J. (2005). West Nile

virus in the vertebrate world. Archives of virology 150, 637-657.

van Marie, G., Antony, J., Ostermann, H., Dunham, C., Hunt, T., Halliday, W.,

Maingat, F., Urbanowski, M. D., Hobman, T., Peeling, J. & Power, C.

(2007). West Nile virus-induced neuroinflammation: glial infection and

102

capsid protein-mediated neurovirulence. Journal of virology 81, 10933-

10949.

Van Waes, C. & Carey, T. E. (1992). Overexpression of the A9 antigen/alpha 6

beta 4 integrin in head and neck cancer. Otolaryngologic clinics of North

America 25,1117-1139.

Vazquez-Ortiz, G., Ciudad, C. J., Pina, P., Vazquez, K., Hidalgo, A., Alatorre, B.,

Garcia, J. A., Salamanca, F., Peralta-Rodriguez, R., Rangel, A. &

Salcedo, M. (2005). Gene identification by cDNA arrays in HPV-positive

cervical cancer. Archives of medical research 36, 448-458.

Vercellotti, G. M. (1990). Proinflammatory and procoagulant effects of herpes

simplex infection on human endothelium. Blood cells 16, 209-215;

discussion 215-206.

Vestweber, D. (2007). Adhesion and signaling molecules contrOlling the

transmigration of leukocytes through endothelium. Immunological reviews

218, 178-196.

Visseren, F. L., Bouwman, J. J., Bouter, K. P., Diepersloot, R. J., de Groot, P. H.

& Erkelens, D. W. (2000). Procoagulant activity of endothelial cells after

infection with respiratory viruses. Thrombosis and haemostasis 84,319-

324.

Vorbrodt, A. W., Dobrogowska, D. H. & Tamawski, M. (2001). Immunogold study

of interendothelial junction-associated and glucose transporter proteins

during postnatal maturation of the mouse blood-brain barrier. Journal of

neurocytology 30,705-716.

103

Wachtel. M .• Frei. K .• Ehler. E.. Fontana. A.. Winterhalter. K. & Gloor. S. M.

(1999). Occludin proteolysis and increased permeability in endothelial

cells through tyrosine phosphatase inhibition. Journal of cell science 112 (

Pt 23).4347-4356.

Wadei. H .• Alangaden. G. J .• Sillix. D. H .• EI-Amm. J. M .• Gruber. S. A.. West. M.

S .• Granger. D. K .• Garnick. J .• Chandrasekar. P .• Migdal. S. D. & Haririan.

A. (2004). West Nile virus encephalitis: an emerging disease in renal

transplant recipients. Clinical transplantation 18.753-758.

Wang. T .• Scully. E.. Yin. Z .• Kim. J. H .• Wang. S .• Van. J .• Mamula. M .•

Anderson. J. F .• Craft. J. & Fikrig. E. (2003a). IFN-gamma-producing

gamma delta T cells help control murine West Nile virus infection. J

ImmunoI171.2524-2531.

Wang. T .• Town. T .• Alexopoulou. L.. Anderson. J. F.. Fikrig. E. & Flavell. R. A.

(2004). Toll-like receptor 3 mediates West Nile virus entry into the brain

causing lethal encephalitis. Nature medicine 10. 1366-1373.

Wang. T. D .• Wang. V. H .• Huang. T. S .• Suo T. C .• Pan. S. L. & Chen. S. V.

(2007). Circulating levels of markers of inflammation and endothelial

activation are increased in men with chronic spinal cord injury. Journal of

the Formosan Medical Association = Taiwan yi zhi 106. 919-928.

Wang. V .• Lobigs. M .• Lee. E. & Mullbacher. A. (2003b). CD8+ T cells mediate

recovery and immunopathology in West Nile virus encephalitis. Journal of

virology n. 13323-13334.

104

Weiss, D., Carr, D., Kellachan, J., Tan, C., Phillips, M., Bresnitz, E. & Layton, M.

(2001). Clinical findings of West Nile virus infection in hospitalized

patients, New York and New Jersey, 2000. Emerging infectious diseases

7,654-658.

Wrtz, I. P. (2008). The selectin-selectin ligand axis in tumor progression. Cancer

Metastasis Rev.

Wong, D., Prameya, R. & Dorovini-Zis, K. (1999). In vitro adhesion and migration

of T lymphocytes across monolayers of human brain microvessel

endothelial cells: regulation by ICAM-1, VCAM-1, E-selectin and PECAM-

1. Joumal of neuropathology and experimental neurology 58, 138-152.

Wunschmann, A., Shivers, J., Carroll, L. & Bender, J. (2004). Pathological and

immunohistochemical findings in American crows (Corvus

brachyrhynchos) naturally infected with West Nile virus. J Vet Diagn Invest

16,329-333.

Xiang, J., Ennis, S. R., Abdelkarim, G. E., Fujisawa, M., Kawai, N. & Keep, R. F.

(2003). Glutamine transport at the blood-brain and blood-cerebrospinal

fluid barriers. Neurochemistry intemational 43, 279-288.

Yang, Y., Estrada, E. Y., Thompson, J. F., Liu, W. & Rosenberg, G. A. (2007).

Matrix metalloproteinase-mediated disruption of tight junction proteins in

cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor

in focal ischemia in rat. J Cereb Blood Flow Metab 27, 697-709.

Zhang, W., Yue, B., Wang, G. Q. & Lu, S. L. (2002). Serum and ascites levels of

macrophage migration inhibitory factor, TNF-alpha and IL-6 in patients

105

with chronic virus hepatitis B and hepatitis cirrhosis. Hepatobiliary

Pancreat Dis Int 1, 577-580.

Zheng, M., Lu, R., Che, X., Li, J., Zhou, C., Wang, L., Xu, Q., Cao, H., Li, Q. &

Yao, Z. (2006). Tyroservatide therapy for tumor growth, invasion and

metastasis of Lewis lung carcinoma and human lung carcinoma A549.

Oncology 70, 418-426.

106