123
Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in Partial Fulfillment of the requirements for the Degree Master of Science Approved November 2013 by the Graduate Supervisory Committee: Jorge Reyes del Valle, Chair Brenda Hogue Wayne D. Frasch ARIZONA STATE UNIVERSITY December 2013

Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

Expression of Dengue virus envelope glycoproteins using

a Measles vaccine vector

by

Rowida Abdelgalel

A Thesis Presented in Partial Fulfillment

of the requirements for the Degree

Master of Science

Approved November 2013 by the

Graduate Supervisory Committee:

Jorge Reyes del Valle, Chair

Brenda Hogue

Wayne D. Frasch

ARIZONA STATE UNIVERSITY

December 2013

Page 2: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

i

ABSTRACT

In terms of prevalence, human suffering and costs dengue infections are the most

important arthropod-borne viral disease worldwide. Dengue virus (DENV) is a mosquito-

borne flavivirus and the etiological agent of dengue fever and dengue hemorrhagic fever.

Thus, development of a safe and efficient vaccine constitutes an urgent necessity. Besides

the traditional strategies aim at generating immunization options, the usage of viral

vectors to deliver antigenic stimulus in order to elicit protection are particularly attractive

for the endeavor of a dengue vaccine. The viral vector (MVvac2) is genetically

equivalent to the currently used measles vaccine strain Moraten, which adds practicality

to my approach. The goal of the present study was to generate a recombinant measles

virus expressing structural antigens from two strains of DENV (DENV2 and DENV4)

The recombinant vectors replication profile was comparable to that of the parental strain

and expresses either membrane bound or soluble forms of DENV2 and DENV4 E

glycoproteins. I discuss future experiments in order to demonstrate its immunogenicity in

our measles-susceptible mouse model.

Page 3: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

ii

TABLE OF CONTENTS

Page

LIST OF TABLES………………………………………………………………........ v

LIST OF FIGURES…………………………………………………………………... vi

ABBERVIATION………………………………………………………….………… ix

CHAPTER

1-INTRODUCTION………………………………………………………………. 1

Dengue viral infections are the cause of major public

health problem worldwide……………………………………………………... 1

Pathogenesis of Dengue infection……………………………………………… 6

Dengue virus genome and virus particles…………………………………….... 9

Dengue virus cycle…………………………………………………………….. 15

Immune response……………………………………………………………… 22

Innate immune response against dengue virus infection……………………… 24

Adaptive immune response against dengue virus infection………………….... 28

Immunopathogenesis of Dengue virus secoundry infection…………..……… 35

Dengue virus vaccine research and developmet progress……………..……… 38

Hypothesis and research specific aims………………………………..………. 50

Page 4: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

iii

The research specific aims………………………………………….….……… 50

Measles virus vector……………………………………………….…...……… 53

2-MATERIAL AND METHODES…………………………………….……..…… 64

Cells and viruses...…………………………………………….…….…....…… 64

Plasmids and plasmids construction…………………………………………... 64

Virus rescue and syncytium formation………………………………………... 68

Virus titration………………………………………………………………….. 72

Virus growth kinetics………………………………………………………….. 74

RNA extraction and RT-PCR………………………………………….……… 74

Western blot assay……………………………………………….………….… 75

Indirect immunofluorescence microscopy……………………………….……. 75

3- RESULT…………………………………………....……………………….….. 77

MVvac2 full-length plasmid vectoring Dengue 2 and 4 glycoprotein………… 77

Reconbinant MV rescue……………………………………………………….. 79

Determination of glycoprotein expression…..………………………………… 83

4- DISCUSSION……………………………………………………..……………. 92

5- REFERENCES……………………………..…….……..……….….…………… 99

Page 5: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

iv

LIST OF TABLES

Table Page

1- Dengue virus vaccines in development………………………………………..…… 41

2- Amplifying primers……………………………………………………….………… 65

3- Sequencing primers.………………………………………………..…….………… 67

4- Mutagenesis primers……………………………………………………….………… 68

Page 6: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

v

LIST OF FIGURES

Figures Page

1- Represent the global distribution of Dengue virus...………………………………… 1

2- Represent the transmission cycles of Dengue………………………………..……… 3

3- Represent the feeding and transmission cycle of mosquito borne Dengue virus….… 5

4- Represent the route of infection of Dengue virus and the consequence of the

infection………………………………………………………………………………… 8

5- Represent the Dengue virus genome and virus particle..………...……….…...…… 11

6- Represent the membrane topology of Dengue virus polyprotein……….………..… 13

7- Schematic representation of the proposed steps of Dengue virus life cycle..……… 18

8- Represent the Initiation of immune response after the inoculation of DENV by

mosquito pit..……………………………………….……………………….………… 25

9- Represention the pathways of DENV antigen representation to effector T cells and T

cell responses to DENV proteins..…………….…………………………….………… 29

10- Represent the mechanisms of neutralization and enhancement by dengue virus-

specific antibodies…………..……………………………………………….………… 32

11- Representation of various Dengue vaccine candidates antigen make up…………. 49

12- Representation of measles virus structure…………………...……………………. 52

Page 7: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

vi

13- Representation of Measles virus replication in the cytoplasm of infected cell..…. 55

14- Represent the relative transcript levels of the six measles genes………………… 56

15- Rescue methods of the Measles virus and recombinant measles virus using standard

cell line……………………..……………………………………………….………… 59

16- representation of the map of (pB+) MVvac(ATU)N……………………………… 61

17- Schematic representation of mutagenesis PCR…………...…………….………… 69

18- Representation of the measles virus rescue strategy.…..………………………… 70

19- Experimental design used in this study……………..………………….………… 73

20- Dengue 4 E protein DIII mutation.……………………………….…….………… 77

21- Map of Dengue virus insect ……………………………….…….………………. 78

22- Restriction endonuclease screening of recombinant MVs plasmid vectoring Dengue 2

coding sequence…………….……………………………………………….………… 79

23- Restriction endonuclease screening of recombinant MVs plasmid vectoring Dengue 4

coding sequence..……………………………………...…………………….………… 80

24- Growth kinetic of cell-associated (Intracellular) and cell-free (extracellular) virus

production in Vero/hSLAm cells.……….………………………………….…….…… 82

25- detection of Dengue 2 PrME and PrMEsol mRNA expression in MVs, Dengue2 virus

and two recombinant MVs infected cells.………………………………….….……… 83

Page 8: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

vii

26- Western blot of protein extract by lysis of Vero h-Slam cells non-infected, infected

with MVvac2 , D2-2 , A.4 or Dengue 2 virus.…..……………….………….………… 84

27- Western blot of protein extract by lysis of Vero h-Slam cells non-infected, infected

with MVvac2 , D2-2 , A.4 or Dengue 2 virus……………………………….………… 86

28-Expression of Dengue2 PrM protein from recombinant MVs. Vero/hSLAM cells. 88

29- Expression of Dengue2 E protein from recombinant MVs. Vero/hSLAM cells….. 89

30- Expression of Dengue4 E protein from recombinant MVs. Vero/hSLAM cells….. 90

31- Expression of Dengue4 PrM protein from recombinant MVs. Vero/hSLAM cells. 91

Page 9: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

viii

ABBREVIATION LIST

Term Definition

WHO World Health Organization

DHF Dengue Hemorrhagic fever

DSS Dengue Shock Syndrome

DF Dengue fever

EPI Extrinsic incubation period

IIP Intrinsic incubation period

C protein Capsid protein

PrM Premature Membrane protein

E protein Envelop protein

PrME Premature Membrane protein and Envelope protein

NS1 Nonstructural protein 1

NS2 Nonstructural protein 2

NS3 Nonstructural protein 3

NS4A Nonstructural protein 4A

NS4B Nonstructural protein 4B

NS5 Nonstructural protein 5

Page 10: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

ix

RNA Ribonucleic acid

mRNA messenger ribonucleic acid

DNA Deoxyribonucleic acid

cDNA complement Deoxyribonucleic acid

ORF Open reading frame

NCR Noncoding regions

Nt Nucleotide

ER Endoplasmic reticulum

N-terminal Amino terminal of protein

C-terminal Carboxyl terminal of protein

TM Transmembrane

Pr Premature

KDa Kilo Dalton

a.a amino acid

sE soluble E protein

DI Domain one

DII Domain two

DIII Domain three

Page 11: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

x

Ig Immunoglobulin

IgG Immunoglobulin G

IgM Immunoglobulin M

IgA Immunoglobulin A

IgE Immunoglobulin E

IgD Immunoglobulin D

INF Interferon

INFβ Interferonβ

INFα Interferonα

INFγ Interferonγ

NTPase Nucleoside triphosphatases

RTPase RNA triphosphatases

RdRP RNA dependent-RNA polymerase

DC-SIGN Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-

integrin

RME Receptor mediated endocytosis

TGN Trans-Golgi network

Ag Antigen

Page 12: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

xi

Abs Antibodies

ATU Additional Transcriptional Unit

NK Natural killer cell

DC Dendritic cell

iDC immature dendritic cell

TNFα Tumernecrosis factor

IL1,2,6,7,8,9 Interleukin 1,2,6,7,8,9

Th1,2,17 T helper cells1,2,17

Tfh Follicular T-cells

TLR Toll like receptor

TCR T cell receptor

BCR B cell receptor

MHC Major histocompatibility complex

ADCC Antibody-Dependent Cell-Mediated Cytotoxicity

C1,3a,5a Complement protein 1, 3a, 5a

PFU plaque-forming unit

CTL Cytotoxic T-lumphocytes

DEN1-4 Dengue virus serotypes 1-4

Page 13: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

xii

MEM modified Eagle Medium

DMEM Dulbecco’s modified Eagle Medium

CPE Cytopathic effect

MOI Multiplicity of infection

TCID50 50% Tissue culture infectious dose

GFP Green Fluorescent protein

JEV Japanese Encephalitis Virus

LAV Live Attenuated Virus

MCS Multiple Cloning Site

MV Measles Virus

MVvac2 Measles Virus vector 2

N protein Nucleocapsid protein

P protein Phosphoprotein

H protein Hemagglutinin

F protein Fusion protein

M protein Matrix protein

L protein L protein

CD 46 Cluster of differentiation 46

Page 14: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

xiii

SLAM Signaling lymphocyte activation molecule

RNP RNA, Nucleocapsid, Phosphoprotein

PCR polymerase chain reaction

IRES Internal ribosome entry site

RT-PCR reverse transcription- polymerase chain reaction

NHP Non- Human Primates

PIV Purified Inactivated Virus

SDS PAGE Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis

SEAR South East Asia Region

WNV West Nile Virus

YFV Yellow Fever Virus

VLP Virus like Particles

Page 15: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

1

Chapter 1-INTRODUCTION

1.1Dengue viral infections are the cause of major public health problems worldwide.

Dengue infection has increased the global health concern. More than 100 countries,

where dengue infection is now endemic, include both tropical and subtropical countries

that have been affected. World Health Organization (WHO) reports “two fifths of the

world's population is at risk of dengue infections with an increasing in the annual number

of cases “(See Figure1). Unfortunately, Dengue causes hospitalization and death mostly

among children. Dengue infection became a very severe problem after 2000 where a

sharp increment in the number of reported dengue infection cases reached 100 million

cases, including one million case of severe dengue infections (dengue hemorrhagic fever,

DHF and dengue shock syndrome, DSS) which lead to 22,000 deaths (6). Generally,

Figure 1, Represent the global distribution of Dengue virus. Dengue virus (DENV) is

endemic in most of the World’s tropical and subtropical areas. The number of dengue fever

DF and dengue hemorrhagic fever DHF cases during one year are shown. Nature reviews

microbilogy(1)

Page 16: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

2

Dengue virus (DENV) infection results in a spectrum of clinical manifestation ranging

from asymptomatic to dengue fever (DF) which is characterized by flu-like-symptoms

(headache, fever and severe eye pain), muscles pain, joint pain, abdominal pain, nausea

and rash (6-8). Common laboratory abnormalities in DF are elevation in serum

transaminases enzymes (indicator of liver damage), thrombocytopenia and leucopenia (9-

12). Full recovery from DF can be expected in most of the cases. Sometimes,

DENVinfections might cause life threatening DHF or DSS. DHF is characterized by all

DF symptoms combined with hemorrhagic tendencies evidenced by spontaneous

bleeding from the mucosa, gastrointestinal tract and other locations, thrombocytopenia

(low platelet count) and evidence of plasma leakage due to increased vascular

permeability (13-15). Characteristically, patients with a high tendency to hemorrhage

present a positive tourniquet sign as evidence for capillary hemorrhage. DSS is

characterized by all DHF symptoms combined with rapid or weak pulse and narrow pulse

pressure defined as the difference between systolic and diastolic blood pressure.

Hemodynamically, the shock syndrome is associated mainly to a reduction in peripheral

resistance caused by a generalized vasodilatation (13, 14). These severe dengue infections

might result in death mostly among patients younger than 15 years old. The WHO

classified DHF in four grades from I to IV. DHF grades I and II represent mild cases

without hemorrhagic manifestations or shock and high number of recovering cases.

Grades III and IV, cases are more severe and are characterized by hemorrhage leading to

shock. The life-threatening DSS stage occurs in the early stage of grade III cases. If the

patients do not receive prompt and appropriate treatment, severe cases of DHF and DSS

Page 17: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

3

may be lethal within 12 to 36 hours after the beginning of a grade IV DHF (16).

DENV is the most important arbovirus (anthropoid –borne) disease that is transmitted to

human by mosquito bites. Anthropoid vector includes Aedes.aegypti, which is the

predominant vector, and Aedes.albopictus, which can sustain transmission in some

endemic areas (17). Aedes mosquitos are the main cause of the disease spreading

worldwide, including South America, North America, Africa, Asia and Australia.

Mosquitos are infected when it imbibes virus with blood meal from an infected host.

After the ingestion of infectious blood meal, the virus infects and multiplies in midgut

Figure 2. Represent the transmission cycles of Dengue virus. Enzootic or sylvatic cycle

takes place in Asia forests has a minimal contribution to the urban epidemic cycle. The

amplification of Dengue virus in the human body result in generating high titer virus in the

blood of infected human which is enough to transmitting virus to mosquito to maintain the

virus through urban epidemic cycle with no need to the enzootic cycle. Which make the

enzootic cycle has no impact in the widespread of Dengue virus in the human population

comparing to other arboviruses such as yellow fever virus where the enzootic cycle contribute

to the widespread of the virus among the human population. Nature Reviews Microbioloy(1)

Page 18: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

4

epithelial cells (18). The virus amplification happens in other mosquito’s tissues and

cells, which results in the distribution of the virus through the hemocele (the mosquito

circulatory system). After the amplification of the virus, it will infect the epithelial cells

of the salivary gland and the mosquitos will be capable of transmitting the virus in the

infected salivary fluid to humans by feeding. The time period from the infection of a

mosquito from an infected host to when the salivary glands became infected and the virus

can be transmitted is called “extrinsic incubation period” (EIP). That is takes about 8 to

12 days depending on the temperature (see figure 3) (18). Mosquitoes are cold blooded

and their metabolism and biology are greatly affected by environmental conditions.

Temperature is one of the most important factors in the viral transmission by mosquitoes.

The temperature directly affects the viral replication in the mosquitoes’ cells and the

increment in temperature increases the rate of the virus replication. However, the

mortality rates of mosquitoes increase at high temperature. The effects of temperature on

mosquitoes and viral replication, may explain the distribution of the DENV infection and

vector in the tropical and subtropical countries, since they have suitable environmental

conditions for the virus replication and for the mosquitos to thrive (18). The disease

control is cumbersome since the geographic expansion of the vector has increased the

circulation of viruses worldwide. Prior to the 1970s, DHF was only reported in five

countries located in South East Asia (13). However, now DHF has been reported in more

than 60 countries and DENV is endemic in more than 100 countries. This dramatic

increase in Dengue infection is due to the geographical expansion of Ae. aegypti after the

vector control efforts decline. Other arboviruses required an enzootic cycle (non-human

Page 19: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

5

primates-mosquito-non-human primates) for virus amplification and maintenance of virus

transmission in human (19). In contrast, DENV is the only virus among the arboviruses

that can be amplified and maintained by urban epidemic transmission cycle (human-

mosquito-human). Enzootic cycle of Dengue transmission does exist in the forests of

Asia, but the contribution of this cycle to the urban epidemic cycle is minimal (see figure

2) (20). Which make the control of the virus transmission by destroying enzootic cycle is

not an option for DENV. Once the infected mosquitoes transmit the virus to a healthy

human host, the virus will replicates into the resident cells at the site of mosquito bite and

the virus will be amplified and spread to infect other tissue and organs (See below). The

time between a human being infected and the onset of symptoms due to infection known

as “intrinsic incubation period” (IIP) ranges from 3 to 14 days, but it generally 4 to 7 days

Figure 3, Represent the feeding and transmission cycle of mosquito borne Dengue

virus. In host #1 high titer virus in the blood occurs at levels that can infect mosquitoes

for 2 to 6 days. After a mosquito ingest virus with blood meal, it takes 8 to 14 days for the

virus to infect the slavery glands, by that time the mosquito became infected and can

transmitted the virus when bite host#2. This period called Extrinsic incubation period

(EIP). The time it takes from infection of host#2 to the onset of symptoms is called

intrinsic incubation period(IIP) in host and it takes 4 to 7 days.

Page 20: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

6

(see figure 3).

1.2 The pathogenesis of Dengue infection depends on the cell and tissue tropism of

DENV. DENV is transmitted by mosquito bites that make the human body’s skin the

entry route of the virus (20). After mosquito bites DENV will pass the epithelial cells

surface where it will reach the subepithelial tissues (21, 22). As a result, the virus will

infect the lymphatic cells that are present in the subepithelial tissues. Epidermal dendritic

cells, known as Langerhans cells, are the main targets of infection; these immature

dendritic cells have a great phagocytic capacity. DENV infection induces

maturation/migration of these cells. The infection of these dendritic cells will cause

lymphatic spread of DENV (23). The infected dendritic cells will migrate to the lymph

nodes where monocytes and macrophages are recruited in which they will be also

infected by DENV(24-27). The infected macrophage and monocytes will circulate

through the blood stream, which is known as primary Viremia (this term denotes the

presence of virus infectivity in the blood stream). Once the virus gets into the blood

stream the viral infection will be localized in the other organs (see figure 4). The

secondary viremia reflects the reboots replication of DENVin host cells; its intensity is

about 105 to 106 Dengue infectious units per ml at its peak (28). It is sustained by

infection of blood –derived monocytes, myeloid dendritic cells and tissues macrophages

(including spleen and liver macrophages). By this time Dengue infection will have two

impacts: the immune responses and the replication of the virus in other tissue and organs.

Infected macrophage will cause the infection of liver, spleen and bone marrow stromal

cells (see figure 4). Other organs have tropism for Dengue infection including kidney,

Page 21: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

7

lung and the central nervous system (28-30). Several organs infection by Dengue is one

of the main events associated with DHF and DSS, where the viraemia is generally 10-100

fold greater than in DF (31, 32). Replication of DENV has different effect on different

organs and tissues. For instance, infected liver macrophages will infect both hepatocytes

and Kupffer cells resulting in necrosis and apoptosis in liver (33). As a result, decreases

in the liver function will happen which cause releasing of toxic products, decreasing the

synthesis of coagulation factors and decreasing in the level of platelets. Losing liver

function develops coagulopathy disorder that may result in progressing of Dengue fever

to DHF and DSS. Replication of DENV in bone marrow stromal cells causes suppression

of haemopoiesis (responsible for the formation of all cellular blood components) that may

cause leukopenia and lymphopenia. Furthermore, the increase of the virus load in the

peripheral monocytes causes severe dysfunction of endothelial cells. In conjunction,

severe thrombocytopenia (decreases in the platelets count) and platelet dysfunction,

associated with the eventual loss of the vascular stability, can occur(34, 35). Thus,

increasing capillary fragility and gastrointestinal mucosal bleeding will take place, which

are characteristic of DHF. In addition, severe infection of endothelial cell causes plasma

leakage, which is characteristic of DSS (13).

Page 22: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

8

Figure 4, Represent the route of infection of Dengue virus and the consequence of the

infection. Mosquito carry dengue virus will bite the skin of the patient result in the

transmission of dengue virus particles into the body. The virus will infect immature

Langerhans cells, which will cause lymphatic spread of the infection and the primary viremia.

The virus will then infect other organs and tissues where the virus will replicate into high load

thus, the secondary viremia will take place and other organs and tissue will be infected

includes brain, lung and kidney. Miedical virology

Page 23: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

9

1.3 Dengue virus genome and virus particles: DENV is a positive single stranded RNA

virus of Flaviviridae family. The DENVencodes structural and non-structural proteins

which includes, capsid (C), Precursor membrane (PrM), envelop (E), nonstructural

protein (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) (see figure 5). DENVcomplex

has four serotypes (Dengue 1-4) (36). Phylogenetically the four Dengue serotypes are

distinct and always cluster by themselves within the mosquito borne flaviviruses branch.

The amino acid sequences of the four serotypes protein are homologous by 63%-68%

while Dengue 3 and Dengue 1 sequence is more related than the other two serotypes (37)

(38). The Dengue mature virus particles has smooth outer surface with icosahedral

scaffold of 90 glycoproteins envelope (E) dimers (39). These 90 E dimers are laid parallel

to the lipid bilayer of the viral membrane where the E proteins are covering the

membrane (M) protein completely in the mature virus. The location of the small M

protein is relative to the overlaying scaffold of E dimers (39). The M protein produced

during the maturation of Dengue virions in the host cells secretory pathway (discussed

later) by the cleavage of the precursor membrane (PrM) protein (40). In contrast, the

outer surface of immature DENV particles are formed with 60 spikes each composed of

three E monomers surrounding a putative trimer of PrM which cover the protruding

fusion peptide in E protein (41) (39). In contrast to the external surface, the virus has less-

well-defined inner isomeric nucleocapsid core which formed by the genomic RNA and

multiple copies of capsid (C) proteins which is surrounded by a host-derived lipid bilayer.

The positive polarity genomic RNA is about 10.6 Kbp with a 5’ type I cap, unlike the

cellular mRNA, DENV genomic RNA lack a 3’ polyadenylate tail (see figure 5) (42).

Page 24: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

10

Genome encodes a single long open reading frame (ORF) flanked by 5’ and 3’ noncoding

regions (NCR). The 5’ and 3’ NCR are about 100 nucleotides (nt) and 700 nucleotides

(nt) respectively and they play a key role in regulation the replication of the genomic

RNA and the translation of the ORF (42, 43). The translation of DENV polyprotein is a

cap dependent and initiated by host cell ribosomal RNA. The large polyprotein will be

directed into the endoplasmic reticulum to be glycosylated by cellular

glycosyltransferases and cleavage into 10 proteins by cellular and viral proteases (see

figure 6). The first proteins to be cleaved are the structural proteins and nonstructural

protein 1 (NS1) as there are a host cellular peptidase between C/PrM, PrM/E and E/NS1.

The DENV encodes serine protease will cleavage between NS2A/NS2B, NS2B/NS3,

NS3/NS4A, NS4A/NS4B and NS4B/NS5. However, the enzyme responsible for cleavage

between NS1/NS2A is unknown (44, 45). The only structural protein to be released from

the polyprotein into the cytoplasm is the capsid (C) protein that is a highly basic α-helical

protein (46). Nascent C (anch C) contains hydrophobic anchor at their c-terminal that

serves as a signal peptide for endoplasmic reticulum (ER) translocation of PrM and the

first signal sequence to be translated by cellular ribosomes. This hydrophobic domain is

cleaved by viral protease to release mature C protein which folded are into compact

dimer with each monomer containing four α helices (47). These will then bind to the

genomic RNA and interact with the membrane surface to form the DENV nucleocapsid

or the viral core. The glycoprotein precursor of M protein contains glycosylation sites at

the N-terminal region, which is down stream of the hydrophobic signal sequence domain

of C protein. A cellular protease called signal peptidase will cleavage downstream the

Page 25: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

11

signal sequence to release the N-terminal of the PrM protein while the C-terminal has the

transmembrane domain (TM) act as ER-retention signals to keep the protein associated

with the ER membrane (4). The major function of the PrM protein is to form a cap to

cover the fusion loop of the E protein to prevent E from fusion during transit through

secretary pathway (40). PrM is about 26 KDa present in the surface of immature virions

which will be cleavage during virus maturation to release the glycosylated,N-terminal Pr

fragment from the small, membrane anchored M protein which is about 9 KDa and

function as scaffold to maintain E proteins folded properly. Envelop protein is Type I

integral glycoprotein and the major protein in the surface of Dengue virions which is

about 53 KDa and mediates host cell receptor binding and membrane fusion (48) (4).

Figure 5, Represent the Dengue virus genome and virus particle. A. the mature Dengue

virion is spherical, enveloped virus contains three structure proteins (Capsid, membrane and

envelop) and RNA genome. The M and E proteins are arranged in the surface of the virion

where the E protein cover the M protein completely. The ectodomain of E protein composed

of three structural or functional domains: Domain I is the central domain(Red), Domain II is

the dimerization domain (yellow) and has the fusion peptide (Green) and Domain III is the

receptor binding domain (Blue). B. The Dengue virus genome is a single stranded, positive –

sense RNA genome. It is capsidated by 5’-type1 structure and doesn't contain polyadenylated

3’-tail. The coding region of the genome is divided into structural and nonstructural protein

and flanked by 3’ and 5’ untraslating regions (UTR). C. The coding regions of the Dengue

genome are shown which is translated into 3 structural proteins and 7 nonstructural proteins.

NMR and X-Ray structures are shown for C, PrM, E, NS3 and NS5.

Page 26: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

12

The E protein is present in two forms metastable or natural pH conformation and

low pH conformation in order to have this flexibility to undergo conformational changes.

E protein has more complex structure compared to the C and M protein (48). E protein

monomer is structurally divided into a two-helix transmembrane region, which is about

50 a.a and also called “steam” and elongated ectodomain . The steam is responsible for

attaching the C-terminal of the E protein into the ER membrane while upstream of the n-

terminal will be cleaved by signal peptidase (49, 50). E protein, lacking the steam region,

will be released from the ER membrane and is called soluble form of E protein (sE). The

ectodomain has three domains that are joint by flexible junction that are capable of hinge-

like motion that are important for the conformational changes of E protein during virions

maturation and fusion of the target cell membrane. Each domain carries out certain

function, Domain I (DI) is flanked by DII and DIII and composed of eight-stranded β-

sheet which participate in the conformational changes induced during viral fusion (39, 50,

51). Domain II (DII) contains hydrophobic fusion loop mediates the virus –cell

membrane fusion at its tip which hidden by the PrM protein the immature virions and by

a hydrophobic pocket that is supplied by DI and DIII of the second E monomer in the

dimer interaction on the surface of mature virions (see figure 6). Domain III has an

immunoglobulin (Ig) like fold and constitute the C-terminal of the E protein ectodomain,

DIII undergoes a major positional rearrangement during cell entry and is proposed as the

receptor binding domain (51). Non-structural protein 1 (NS1) is a translocated

glycoprotein into the ER membrane by the c-terminal signal sequence of E protein and

cleavage from the E protein by host signal peptidase and cleavage from NS2A by

Page 27: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

13

unknown protease. Cleavage NS1 could present in the cell surface in form of highly

stable homodimers or be secreted into the extracellular space. The function of NS1 is not

clear yet, NS1 plays a role in the DENV RNA replication as it was demonstrated that

introducing mutation to NS1 lead to dramatic defects in RNA replication(52). In addition,

NS1 play a key role in the immunopathogenesis of DENV. The function of the non-

structural protein 2A(NS2A) is not clear however, there is some evidence that NS2A can

act as an interferon (IFN) antagonist and assist the DENV evasion of interferon inhibition

of viral replication (53). One of the most important non-structural proteins is NS3 which

carry out multifunction includes serine like protease activity, RNA helicase- NTPase and

RTPase (54). The N-terminal of the NS3 has the protease domain (180 a.a), which

cleaves the viral protein at multiple sites (see figure 6, discussed above).

Figure 6, Represent the membrane topology of Dengue virus polyprotein. The viral RNA

is translated as a polyprotein and processed by cellular and viral proteases (represented by

arrows with different colors). The structure proteins are released from the poly proteins by

signalase cleavage in the ER. The PrM and E proteins remain anchored in the lumen side of

the ER membrane and the C protein remain anchored in the cytoplasm side of the ER where it

will be cleavage later by the NS2b/NS3 protease. The nonstructural proteins are released from

the ployprotein by NS2B/NS3 protease in the cytoplasm with exception of NS1 which

cleavage by unknown protease on its c-terminal. The C, NS1, NS3 and NS5/NS4Bwill be

disassociate from the ER membrane while the other protein will be membrane anchored. Cell

(4)

Page 28: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

14

The protease activity of the NS3 is dependent on the presence of the co-factor

NS2B which will form an NS2B-NS3 serine protease complex (45). The cofactor NS2B

warps around the protease domain of NS3 in a belt-like structure and anchored its

hydrophobic region into the ER membrane to bring the protease domain close the

transmembrane domain that must be cleaved within the polyprotein (38) . The C-terminal

region of the NS3 protein has nucleic acid modifying activates which are essential for the

Viral RNA replication. RNA helicase-NTPase is an enzyme has RNA-stimulated

nucleoside triphosphatase (NTPase) activity and RNA unwinding activity. The helicase-

NTPase domain is composed of three subdomains, where two of them represent the

NTPase activity and the third part carry out the RNA unwinding activity . NS3 C-

terminal encodes an RNA triphosphatase activity (RTPase) responsible for

dephosphorylate the 5’end of the viral RNA before cap addition . NS4A and NSB are

small hydrophobic proteins which act as antagonist to block type I INF signaling

pathways that are essential to enhance the DENV replication (53). The largest (104KDa)

and the most conserved protein among Dengue serotypes is the non-structural NS5

protein. NS5 is a multifunctional protein with methyltransferase (MTase) and RNA

dependent-RNA polymerase (RdRP) activities (55). The N-terminal of the NS3 protein

has the MTase domain, which involves in the modification of the Viral RNA 5’ cap. The

C-terminal of NS5 contains the RdRp essential for the RNA replication. In addition to its

role in the RNA replication, NS5 has the ability to block the signaling pathways of type I

interferon to enhance the viral replication and infectivity (56, 57).

Page 29: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

15

1.4 Dengue virus infectious cycle: The basic stages of the DENV life cycle include;

attachment to cell surface, internalization into the host cell, releasing of the nucleocapsid

and transfer of RNA into cytoplasm, translation of the viral protein, replication of

genomic RNA, assembly and maturation of the virions, and ultimately the release of

mature viruses from the cells (see figure7)(36, 40). The early stages in the DENV life

cycle involve the viral entry, which is mediated by receptor recognition and attachment

(see figure 7 a.1). E glycoprotein is responsible for the receptor recognition and

attachment to concentrate the virions in the surface of host cells. The recognition of virus

–specific cell receptor contributes to host range and tissue tropism (the tissues where the

virus can replicate) (36). DENV has a wide range of tissue tropism and host range of

human and mosquitoes that is why it is unclear to determine the virus’s specific cell

receptor is unclear. Dendritic cells have a surface protein called DC-SIGN (Dendritic

Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) that is the

putative primary protein receptor for DENV (58, 59). The attachment and the

concentration of the virus particles on the surface of the host cells is mainly mediated by

the domain III of E protein (60-62). However, there is a differential receptor usage by the

four Dengue serotypes. For instance, DENV-1 specific binding protein on human liver

cells is the 37/67KDa high-affinity laminin receptor (63), while the GRP78 glucose-

regulated protein has been identified as a receptor for DENV-2 internalization into human

liver cells (64, 65). The glycosylation pattern on the viral surface varies among different

Dengue serotypes that can explain the differential receptor usage among these serotypes.

In addition to the viral factors, cell factors such as heat sock protein associated with

Page 30: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

16

membrane micro-domains or lipid rafts can participate in DENV entry as a receptor

complex which includes Hsp90 and Hsp70 in human monocytes and macrophages as well

as gp45 and 74 KDa proteins in mosquito cells (66, 67). The internalization of DENV

into the host cells happen by receptor-mediated endocytosis (RME), also called clathrin-

dependent endocytosis, is a process by which cells internalize (endocytosis) the virus

particles by the inward budding of plasma membrane vesicles containing the E proteins

attachment with surface receptor (see figure 7 a.2) (68, 69). The plasma membrane

vesicles bud into the cytoplasm and deliver the virus particle to early endosomes. Early

endosomes mature in several ways to form late endosomes as they become increasingly

acidic at which the virus fusion occurs (see figure 7.b). Flaviviruses are considered

important models for studying viral membrane fusion as they exhibit the most efficient

and the fastest fusion machinery of all enveloped viruses (48). DENV membrane fusion

is mediated by a class II viral fusion protein, the major enveloped protein E has

hydrophobic sequence represent the fusion peptide which mediate the fusion of the viral

lipid membrane with the endosome lipid membrane (70). The initiation of the fusion

processes depend on some factors that include: acidic pH, lipid compassion of the target

membrane, and histidine residues in the E protein (48).

Page 31: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

17

Page 32: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

18

Figure 7, ( Previous page) Schematic representation of the proposed steps of Dengue virus

life cycle: a. Attachment and receptor mediate endocytosis of the mature Dengue virion.

Color codes; Dengue virus nucleocapsid is colored gray , Dengue virus E protein DI, DII and

DIII are colored red, yellow and blue, respectively and the transmembrane domain is colored

green. The outer/inner leaflets of the viral lipid membrane and the host cell membrane are

colored yellow/blue and gray/red. The target cell receptor is colored orang. a.1. shown the

attachment of the virion to specific cell receptor. The surface of the mature virion formed by

90 homo-dimers of E protein in a metastable form where the D III of E protein will attach to

the receptor. a.2. show the internalization of Dengue virion into the host cell by receptor

mediated endocytosis (RME) where the cell internalize the virus by inward budding of plasma

membrane. b.pH dependent fusion of the virus membrane with the endosome membrane

and the releasing of the virus nucleocapsid. Color codes; the same as part a of this part,

Fusion peptide at the distal end of DII is indicated in orange. b.1. low endosomal pH induce

the outward extension the steam region, followed by the dissociation of E dimers into

monomer ,expose the fusion loop and the interaction of the fusion loop with the endosomal

target membrane. b.2. the formation of E trimer and the protonation of histidine residues

induce the back-folding of DIII and Zipping-up of the steam. b.3. Hemi-fusion intermediate

the formation of fusion pore by mixing the outer leaflets of the virus and the endosome

membrane. b.4. generation of the final post-fusion structure (E trimer and the back-folding of

E protein DIII and zipping up the steam) and opening of fusion pore. b.5. the pore in the virus

and endosome membrane allow the releasing of the nucleocapsid includes the core protein and

the genomic RNA which will be transfer into the cytoplasm where it will be in a close contact

with the ER. c.Translation and replication of the genomic RNA. The translated polyprotein

is directed into The ER while it is attached to the ribosomes. The RNA dependent RNA

polymerase will be released from the polyprotein which will carry out the replication of the

genomic RNA in the cytoplasm in close contact with ER. Some of the protein will be released

from the polyprotein and the ER and the rest will be membrane anchored in the ER (for details

see figure #). d.virus assembly in the ER and maturation in the TGN. d.1. Virus assembly

takes place when there are enough envelop protein displayed in membrane of the ER where

the E protein exists as heterodimer with PrM, viral protein then and the nucleic acid combine

to form virus particles which will gain lipid membrane by biding from the ER. d.2. After

assembly the immature virions transit from the ER to the Golgi network where the virions

encounter a natural pH. The immature virions under a natural pH have a spiky surface. The

arrangement of the E protein (gray) in 60 trimeric of heterodimer with PrM protein (Pr peptide

represented by blue color), which extend away from the virions surface gave the virions the

spiky surface morphology. d.3. during transportation through Trans Golgi Network (TGN) the

virions encounter acidic pH which initiated conformational changes of the surface proteins.

The PrM-E heterodimers dissociate from their trimeric spike like organization and form 90

dimers that lie flat against the virus surface. d.4.the maturation of the virions include cleavage

of the Pr peptide by cellular furin protease where the Pr peptide maintains its position as a cap

of E protein(Pr+M-E) as the virions transit through TGN and cellular secretory pathways. e.

Mature virus releasing. Following furin cleavage, the mature virions is secreted is secreted

into extracellular space and the Pr peptide is released from the mature particle where the E

proteins remain as 90 homodimers lying parallel to the virions surface and M protein is

beneath the E protein shell and embedded in to the viral membrane.

Page 33: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

19

The acidic environment on the late endosomes is the key activator of the E protein

fusion activity that is known as pH dependent fusion. The pH of 6.6 and lower is suitable

to activate the fusion process. However, the exposure of the virus alone to acidic pH

result in inactivation of its fusion ability, which suggests that lipid membrane, plays a role

in the fusion activity of DENV E protein(48).

The lipid composition of endosome membrane influence the fusion efficiency, for

instance, cholesterol –rich lipids rafts can enhance the internalization and the fusion of

DENV (71). Viral factor can also contribute in the initiation of the fusion, as the histidine

residues in the DENV E protein act as sensors of pH that trigger conformational changes

in the E protein (72). At natural pH the histidine residues are un-charged but at the acidic

pH they became positively charged by the addition of extra protons. The protonation of

histidine residues could release D III of E protein to permit folding-back conformation

and hairpin formation that is necessary for the formation of fusion pour (72). Up on the

endocytosis of DENV, the pre-fusion E protein at a natural pH is on its metastable state of

homodimers parallel to the lipid membrane of the DENV. One of the requirements of

endosome trafficking and maturation in the living cells is generating an acidic

environment inside the endosome vesicles (48). Once the pH riches 6.6 or lower, the

protonation of the histidine residues on the E protein takes place (73). Which trigger the

E protein to undergo conformational and positional transformation by the outward

extension of the stream region, accompanied by the dissociation of E dimer into

monomer (see figure 7 b.1) (74). The dissociation of E homodimer results in the exposure

of the hydrophobic fusion loop. As a consequence of all these motions, the fusion loop

Page 34: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

20

will interact with the lipid membrane of the endosome. In order to establish a close

positioning with target membrane, the viral E protein arranges in a timers where the E

monomers are parallel to one another and perpendicular to the target membrane (72). The

fusion loops are exposed on one end of the timer (figure7 b.2). The DIII of the E protein

undergoes major conformational changes where it folds back to bring the steam sequence

in the viral envelop to be in close contact to the fusion loop in the tip of the DII (75). As a

consequence, the viral membrane will be very close to the target membrane and a hemi -

fusion intermediate the formation of fusion pour by mixing the outer leaflets of the viral

and target membranes (figure 7 b.3) (76). Finally the fusion pour formed and maintained

because of the back-folding of E protein DIII and the zipping up of the steam region

(figure 7 b.4) (72). The fusion pour in the endosome membrane allow the release of the

nucleocapsid into the cytoplasm. Then the RNA is transferred into cytoplasm to be

replicated (figure7 b.5). Initially, the released viral RNA is translated into a polyprotein,

which then is directed into the endoplasmic reticulum (ER) (figure 7 c.). Signal sequence

within the poly protein translocate the E, PrM and NS1 protein into the lumen of the ER,

while the C protein, NS3 and NS5 are released into the cytoplasm (72). The process of

this polyprotein releases the NS5 that contains the RNA dependent RNA polymerase

(RdRp), which must happen before the RNA replication. Genomic RNA replication

begins with the synthesis of a genome-length minus stranded RNA, which then serve as a

template for the synthesis of additional plus stranded RNA(77). The viral RNA

replication is asymmetric where the positive strands accumulate ten times more than the

minus strands (78). Virus assembly takes place once there is enough envelop protein

Page 35: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

21

displayed in the membrane of the ER (figure 7d.1). The assembly occurs in the ER when

the newly synthesized viral protein and nucleic acid combine to form immature –fusion-

incompetent virus particles (78). After assembling, the virions transit from the ER

through trans-Golgi network (TGN) to the cell surface. Budding assembles the virions

from the ER, which is responsible for the gain of host derived lipid envelope. Through

this trip the immature virions undergo a protease- and pH- dependent maturation (79, 80).

During the maturation, E proteins go through positional reorganization, while the PrM

protein is cleaved by cellular furin protease. The conformational changes in E protein are

triggered by the low pH in the exocytosis vesicles and occur before the cleavage of the Pr

peptide by the furin protease (81). The structural change from the immature virions with

spiky surface to mature virions with smooth surface occurs during transportation through

the TGN (figure7, d.2 and d.3). The initial bud of immature virion into the ER happen

under a natural pH environment where the E protein form a 60 trimeric spikes that

extends away from the surface of the virions where the “ Pr ” peptide cover each E

monomer (figure 7,d.1). During its transit through the secretory pathways, the virions

encounter acidic pH in the TGN. Under the acidic condition the PrM-E protein

heterodimer disassociate from the trimeric spike- like organization to form dimers of

PrM- E protein which lie flat against the viral surface, forming the 90 dimers envelop

shell of DENV. This rearrangement of E protein gives the virions its smooth surface

morphology (figure7 d.3) (39, 60). As the virions transit through the TNG, The cellular

furin protease cleavages the glycosylated-N-terminal Pr peptide from the small-

membrane anchored M protein that will remain associated with the virions. However, the

Page 36: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

22

cleaved Pr peptides remain in its position as a cap on E protein during the trafficking in

the TNG (figure7, d.4) (60). Once the virions are released to the extracellular space the Pr

peptide will be released and the virions will be on its mature, infectious form where the E

protein arranged in a 90 homodimer lies against the virus membrane and completely

cover the small M protein (figure7,e.). An interesting aspect of the DENV life cycle is the

generation of subviral particles that contains only the PrM and E protein that are

associated with the lipid membrane without the nucleocapsid. These particles can be

presented as natural by-product in the supernatants of infected cells. These particles are

slightly less dense than the virions, they are 30 nm in diameter and are called

recombinant subviral particle (RSP) (36, 60).

1.4. IMMUNE RESPONSE: Immunity is the collective and coordinated actions taken

by cells and molecules of the immune response toward a foreign substance such as a viral

infection. Immunity has two lines of defense early defense called innate immunity

(known as native immunity as well) and late immunity called adaptive immunity. Each

defense line has its own cells and molecules. Different effector cells mediate innate

immunity such as phagocytic cells, which recognize the microbial substance or the

antigen (Ag) at the site of infection, ingest and destruct the ingested microbe. Phagocytic

cells include macrophages and dendritic cells which play a key role in linking innate and

adaptive immunity by presenting the antigen to adaptive immunity cells. Granulocytes

have inflammatory granules in their cytoplasm and are composed of three types:

neutrophils, eosinophils and basophils(82). Natural killer cells (NK) are bone marrow

derived lymphocytes which mediate cytolysis of virus infected cells to clear the viral

Page 37: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

23

infection. Cytokines are a large and heterogeneous group of secreted proteins produced

by many different cells of innate immunity and adaptive immunity later on. Cytokines

have different categories depending on their biological actions.

Proinflamatory cytokines are secreted proteins that promote activation of immunity cells

and they include, α-Interferon (INFα), β-Interferon (INFβ), tumoral necrosis factor

(TNFα), IL6, IL2 and INF-δ. Chemokines are secreted proteins that act as chemo

attractants to recruit immune cells to the site of infection such as CXCL-8, CXCL-3 and

CXCL-1. Complement proteins are a group of more than 30 different plasma proteins that

interact with each other and bind with the pathogen to kill the pathogen. Adaptive

immunity is composed of the humoral and cellular immune response. Antibodies are Y

shape proteins where the two arms of the antibodies recognize identical antigen sites.

Antibodies are secreted by lymphocytes cells called B-lymphocytes which secret Ab to

the extracellular space or display the Ab on their surface (82, 83). Cell mediated

immunity (also known as cellular immunity) is mediated by T-lymphocyte cells which

divide into two major categories: Helper T-lymphocytes (Th) and Cytotoxic T-

lymphocytes (CTL). They have different effector functions and different surface markers.

Helper T-lymphocytes carry a surface protein called CD4 and have at least four main

types of CD4+ effector T-cells called Th1, Th2, Th17 and follicular helper T-cells (Tfh).

Each one of these subclasses promotes different immune responses depending on the type

of infection. Cytotoxic T-lymphocytes carry a surface protein called CD8 and are

responsible for killing infected cells that display the antigen on their surface. Innate and

adaptive Immune cells are localized and generated from anatomically identified tissues

Page 38: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

24

and organs. Lymphoid tissues and organs divide into two major types: Central (primary)

lymphoid is the generative organs and tissues and the Peripheral (secondary) lymphoid is

the site where antigens present to the naïve immune cells in order to be activated.

Includes spleen and lymphoid nodes that distribute around the body and are connected

through the lymphatic system where the immune cells and molecules are circulated in a

resting state until they get activated (82, 83).

1.4.1. Innate immune response against dengue virus infection has two phases; the

immediate phase of inhibition of DENV infection by type I Interferon pathway and early

phase of elimination of DENV infection by NK cell-mediate killing of infected cells.

One of the most remarkable pathways of host cell defense to limit the viral infection is

type I interferon system. A few hours after the initial bite of an infected mosquito and

prior to recruitment of other immune cells the Interferon response will be activated (see

figure 8) (84, 85). Type I interferon is induced by almost any type of cells in response to

DENV infection but some cells seems to be specialized for the task. These are immature

dendritic cells (Langerhans cells), which are considered the first innate immune cells to

be infected with DENV (see figure 8). There is two classes of antiviral type I interferon:

INF-α and INF-β that are induced by cytoplasmic sensors of Dengue viral RNA, RIG-I

(retinoic acid inducible gene 1) and MDA-5 (Melanoma Differentiation – Associated

protein-5) in the cytoplasm of infected cells will trigger also the secretion of type

interferon upon recognition of RNA motifs (86). Production of INF-α/β will affect the

infected and uninfected cells, as they will bind to interferon receptors in the surface of

these cells. The interferon receptors will activate intercellular cell signaling pathways that

Page 39: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

25

aim to stimulate the transcription of more than 100 different genes with antiviral

activities. One such protein is the dsRNA–dependent protein kinase called PKR that

inhibits the translation of the viral protein and inhibits viral replication (87). However,

DENV has evolved mechanisms to escape from the interferon response. These

mechanisms are the inhibition of both the production and signaling of type I INF. Non-

structural proteins NS2A to NS5 responsible for DENV evasion (86). For instance, the

NS2B3 protease complex functions as an antagonist to block the INF receptors. NS2A,

Figure 8. Represent the Initiation of immune response after the inoculation of DENV by

mosquito pit. Infection by dengue virus (DENV) occur after subcutaneous injection of the

virus into the skin. Released Dengue viral particles may infect nearby cells (thought to be

predominantly Langerhans cell or dendritic cells (iDCs)). The infection of iDC activates

resident immune cells includes granulocytes such as mast cells which will initiate

inflammatory response. A local inflammatory response to DENV in the skin prompts the

recruitment of leukocytes from the vasculature, including natural killer (NK) cells and T cells,

which promote the killing of virus-infected cells at the site of injection. At the main time,

infected dendritic cells and macrophages travel to draining lymph nodes via lymphatic vessels

to establish systemic infection as described in FIG 4. Nature Reviews(3)

Page 40: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

26

NS4A, NS4B and NS5 function as antagonists of type I IFN signaling by targeting

different components of the signaling pathways. INF-α/β play an important role in the

mechanisms of host defense against DENV but, once the viral replication has been

established the INF system has little effect on DENV. At the same time, DENV

replications in immature dendritic cells (Langerhans cells) are facilitated by the

internalization of DENV through the adhesion of the E protein with cellular receptor such

as Dendritic Cell-Specific Intracellular adhesion molecule-3-Grabbing Non-integrin (DC-

SIGN) (see figure 8) (59, 88). It has been observed that a dramatic inhibition of DENV

infection happens by blockade of DC-SIGN receptors, thus DCs play a central role in the

establishment of DENV infection and stimulation of a strong immune response. The

normal function of the Langerhans cells is to capture antigens and process them into

immunogenic peptides, migrate from the site of infection to the nearby lymph node to

present the antigen to other lymphocytes. Inside the Langerhans endosomes there are

sensors called Toll Like Receptors (TLR), which recognize DENV ligands, derived from

its life cycle. TLR molecules are conserved proteins and there is 11 types of TLR that

have been identified in humans. TLR molecules that involve in DENV recognition are

TLR 7, TLR 8 and TLR 9 (89, 90). They trigger the production of cytokines such as IL-2,

IL-12, IL-6, TNFα, and INFα/β. All this cytokines set up a state of inflammation in the

tissue that is associated to dilation of local blood vessels and changes in the endothelial

cells of their wall. These changes happen prior to the recruitment of other immune cells

and lead to the movement of leukocytes such as neutrophils and monocytes out of the

blood vessel and into the infected tissue. The blood vessels become more permeable

Page 41: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

27

which will facilitate the migration of other leukocytes and lymphocytes to the site of

infection (see figure 8). Once the monocytes migrate into the tissue they will

differentiate into macrophages. Neutrophils, non-infected Langerhans and macrophages

will recognize, ingest and destroy DENV, which present in the extracellular space.

Activated macrophages and Langerhans cells will secret cytokines such as IL1-β which

activates lymphocytes and increase access of effector cells, TNF-α which increase

vascular permeability which lead to an increase in the entry of plasma proteins and cells

and fluid drainage to lymph node, CXCL8 which is a chemotactic factor that recruits

Natural killer (NK) to the site of infection (91). Since, DENV is an obligated intracellular

parasites. Natural killer cell-mediated killing of infected cells clears the infection at this

early phase( see figure 8). Gathering of NK cells and Langerhans (iDC) in the inflamed

tissue result is physical contact between NK and Langerhans (iDC) which promotes

events including, Langerhans (iDC)- induce NK proliferation, NK-mediated shattering of

Langerhans (iDC) and NK-dependent Langerhans maturation (92). The conversation

between NK and iDC promotes the quality and the intensity of innate response and

activates appropriate adaptive response against DENV infection. iDC releases cytokines

such as IL-12 and IL-2 which induce the proliferation and activation of NK by releasing

cytokines like IFN-δ and TNFα. Those cytokines will induce the maturation of

Langerhans (iDC) to the mature form of dendritic cells that are characterized by special

surface marker which will migrate to the lymph node. Mature DC releases type I

interferon that increase the cytotoxicity of NK to mediate killing of infected cells

including infected Langerhans cells (iDC). NK-mediated shattering of Langerhans (iDC)

Page 42: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

28

as it expresses DENV antigen on its surface as infected cells which stimulated NK to

destroy it order to prevent the spread of the infection to other lymphocytes in the lymph

node. At this point, dendritic cells and macrophages will migrate to the lymph node for

subsequent presentation of DENV antigen to naïve T and B-lymphocytes to activate the

adaptive immune response (92).

1.4.2. Adaptive immune response against dengue virus infection has two goals: The

blocking of the viral entry by antibodies secreted by B-lymphocytes OR elimination of

infected cells by Cytotoxic T-lymphocytes. Naïve lymphocytes are normally circulating

throughout the body in a “resting” state. Once they become encounter with DENV Ag

from APC (i.e., macrophages and dendritic cells) a series of events will take place; 1-

Lymphocytes activation, 2- clonal expansion or multiplication of lymphocytes, 3-

differentiation into either effector lymphocytes or memory lymphocytes, and 4-migration

from the lymph node to the site of the infected tissue (3). Cellular immune response

mediated by CD4+ and CD8+ T-cells are activated in the lymphocyte through Antigen

presentation. T-lymphocytes have a special cell marker called T cell Receptor (TCR).

This receptor is responsible for recognizing the Ag. Both CD4 and CD8 used TCR to

recognize the Ag but each cell required a different ways of antigen presentation (93).

When the virus enters the cells and start to synthesis their proteins, the newly synthesized

viral protein will be presented through either MHC class I or MHC class II presentation

pathways. MHC molecule has a binding groove where the Ag will be bound and

presented to TCR on the cell surface (see figure 9). MHC class I molecules present

DENV antigen to CD8+ T-cells while MHC class II molecules present DENV antigen to

Page 43: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

29

CD4+ T-cells (3). Both MHC class I and III are displayed on the surface of APC . MHC

class I present in almost all nucleated cells. TCR has the affinity to bind to specific

DENV Ag epitopes where it recognizes antigen epitopes throughout DENV polyprotein,

however the most recognized Ag epitopes present in the NS3 protein (94). DENV

specific T-cells display different effector functions including; targeting cell lysis and

production of a range of cytokines.

Figure 9, Represention the pathways of DENV antigen representation to effector T

cells and T cell responses to DENV proteins, the dengue virus life cycle and sources of

antigen are show. Viral attachment, internalization, fusion and translation proceed as

described in FIG 7. Newly synthesized viral proteins enter MHC class I or II presentation

pathways and viral peptide epitopes are presented on the cell surface within the binding

groove of MHC molecules. MHC class I present DENV antigen peptide to the CD8+ T-

cells which response by lysis target cell. MHC class II present DENV antigen peptide to

CD4+ T-cells which response by cytokines secretion. Both CD8+ and CD4+ T-cells bind

to the peptides, which match the epitopes of their T-cell receptor (TCR). Nature Reviews

Immunology(3)

Page 44: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

30

Effector Functions of DENV specific CD4+ T-cells (Th): a broad range of

cytokines is produced by the helper T-cells (Th) in response to recognition of DENV

antigen peptide-MHC complex on the surface of macrophage and dendritic cells. The

pattern of cytokines production follows certain sub-classes of T-helper cells called T-

helper 1 (Th1) includes INF-δ, IL2 and TNFα. The principal stimulation of Th1 cells is

the intracellular microbes such as DENV. Th1 effector functions include; 1-activation of

macrophage to ingest and destroy DENV, 2-activation of B-cells to secret DENV specific

antibodies, 3- immune response amplification (93). The signature cytokines of Th1 is

IFN-δ, which is a principal macrophage activating cytokines. Once macrophages are

activated by INF-δ they will produce molecules such as reactive oxygen, nitric oxide and

increases the production of lysosomal enzymes, which increase the phagocytosis of

DENV and enhance the killing of ingested viruses (95). Activated macrophages secrete

cytokines required for Th1 differentiation such as IL1, IL12 and TNFα, which also recruit

leukocytes and promote inflammations. INF-δ secreted by Th1 stimulates B-cells to

secrete antibodies. In addition, IL-2 and TNFα secreted by Th1 promote the

differentiation of CD4+T-cells to Th1 subclass and inhibit the other T-helper subclasses

such as Th2 and Th17 in a process called amplification of immune response (3, 96, 97).

Effector function of Dengue specific CD8+ T-cells (CTL): Elimination of DENV

infected cells by cytotoxic mediated killing of the cells. CTL recognize DENV antigen

epitope-MHC class I complex on the surface of infected cells where the CTL will be

activated to secrete cytotoxic proteins including granzymes and perforin which induce

cell apoptosis by cleave cellular proteins such proteins are stored within cytoplasmic

Page 45: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

31

granules in the CTL. Once the cytotoxic ability of CTL is activated a series of events take

place on the cytoplasm of the CTL; 1- microtubule in cytoplasm of CTL moves the

cytoplasmic granules to be concentrated in the region of contact with infected cells, 2-

granules will fuse with the plasma membrane of CTL at the site of connection with the

infected cell. 3-exocytosis of CTL’s granules and detachment of CTL from infected cells.

The death of the infected cells occurs during the next 2 hours and happens even if the

CTL detaches (93, 97-99).

Humoral response against DENV is mediated by secreted antibodies, which are

produced by B-lymphocytes. Resting naïve B-cells have either IgM or IgD

immunoglobulin on its surface that will be used to recognize DENV antigen through the

process of antigen presentation on the lymph node that called the recognition phase. The

proliferation and differentiation of B-cells depend on the stimulation of T-helper cells.

The antibody responses to DENV antigen require that the antigen be recognized and

internalized by B-cell and that peptide of DENV antigen should be presented to the T-

helper cells at the lymph node that will stimulate the B-cell to enter the activation phase

(100). During that phase, B-cells specific to that antigen peptide will be multiplied

through the clonal expansion process and then will be differentiated either to memory B-

cells or to Plasma cells which will secrete antibodies. The activation of B-cells by T-

helper cells, known as helper T-cells dependent antibody response, mainly happens in

response to protein antigen such as DENV proteins antigens (3). Once the B-cells are

activated by T-helper-cells their surface marker will be changed through a process called

isotopes switching (3) (100, 101). As was mentioned earlier, DENV infection activated

Page 46: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

32

the pathways of Th1 cells that promote the heavy chain isotype switching to IgG instead

of IgM and IgD. The secreted Ig main function is mediated by binding to the antigen

through the epitopes of the light chain, but also to different effector cells. Thus, Th1

promotes isotype switching to IgG to clear DENV infection. Antibodies will be secreted

from plasma cells at lymph node and migrate to perform its effector functions at the site

of infection (102).

Figure 10, Represent the mechanisms of neutralization and enhancement by dengue

virus-specific antibodies, A. At high levels of epitopes occupancy, dengue specific antibodies

can block the fusion at a post-binding stage or after the viral entry to prevent the release of the

virus particles from the endosomes. B. At high levels of epitopes occupancy, dengue specific

antibodies can block the binding of virus particles and inhibit the viral entry. C. At low levels

of epitopes occupancy, dengue specific antibodies can enhance the uptake of virus particles in

to cells by the interaction between the Fc receptor on the surface of host cell and the heavy

chain of the IgG in a phenomenon called “antibody dependent enhancement” of the disease.

Nature Reviews Immunology(3)

Page 47: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

33

Effector functions of Dengue virus specific antibodies: Includes neutralization of

DENV (the most direct way to protect host against DENV infection by binding to the

viral envelope proteins and prevent it from entry of host cells). Opsonization is the

coating of DENV by antibodies and subsequent phagocytosis of DENV, complement

activation against DENV and antibody-dependent cellular cytotoxicity (see figure 10)

(103). The main targets of antibody responses to DENV are the precursor membrane

(PrM), envelope (E) structure protein and non-structural protein 1 (NS1) (100, 104).

DENV E glycoprotein is the main surface component of the virus particles and has the

most extensive antibody response. E protein domain III contains the putative receptor-

binding site, which is the binding region that allows the virus to attach to the target cells.

That makes Domain III the most accessible part of E protein and the most variable in

amino acid sequence between serotypes (104). Antibodies specific for domain III are

highly specialized among serotypes. However, they consist a small fraction of the total

antibody in immune sera binding to DENV as it was demonstrated by a study that

neutralized DENV by human immune sera (103, 105). This study proposed that

antibodies directed to other epitopes are responsible for the most of neutralizing of

DENV. M protein is completely hidden by the E protein dimers and is in the mature

virions. In some cases, an incomplete cleavage of the PrM results in partial mature or

immature virions to be released from the cells where the anti-PrM antibodies will

recognize the glycoprotein in the surface of the virions. NS1 protein is produced as an

extracellular protein or presents on the surface of infected cells, thus the antibodies

against NS1 do not play a role in neutralizing of DENV but has a central role in antibody-

Page 48: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

34

dependent cellular cytotoxicity (100). Natural killer (NK) cell has a surface molecules

caller FcδR III which is a receptor for IgG where the NK binds to the heavy chain of the

anti NS1 IgG which cover DENV infected cells in a process called antibody-dependent

cellular cytotoxicity (ADCC) (see figure 10) (3). The NK is activated to kill the antibody-

coated cells. The main function of anti-E antibodies is to neutralize and eliminate DENV.

Anti-E antibodies bind to E glycoprotein and block the ability of DENV to attach or to

enter the target cells. The antigen binding regions of these antibodies are highly

specialized to recognize a certain amino acid sequence. These are known as monoclonal

antibodies and they are different among different DENV serotypes (103). Antibodies

against PrM and E protein mediate opsonization and phagocytosis of DENV.

Macrophages have a surface protein called FcδR I that is a receptor for IgG. The anti-E

coat the surface of Dengue mature virions and anti-PrM coat the surface of immature

Dengue virions. The heavy chain of those IgG antibodies may bind to the FcδR I receptor

on the surface of the macrophages and activate the macrophages to ingest and destroy

antibodies coated (opsonized) Dengue virions. Anti-PrM and E protein coated the

Dengue virions activate the complement system to eliminate the virus from the

extracellular space (106) (100, 107). Complement system composed of plasma proteins

that bind to the heavy chain of the antibodies coating the virions. In the case of DENV

infection, the classical pathway is the most common pathway since IgG is the

dominating. Classical activation of the complement system is initiated by the binding of

complement protein 1 (C1) to the heavy chain of anti-E or PrM IgG followed by a

sequential binding of other complement proteins including C3a and C5a (108, 109).

Page 49: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

35

Activation of the complement system results in the following actions; opsonization of

DENV to macrophages and neutrophils, activation the phagocytosis of the virus, and

stimulation of inflammatory response as the C5a and C3a protein induce inflammation by

activating neutrophils, eosinophils and basophils (109). Although these induction of the

inflammation trigger plasma leakage of endothelial cells which result in unfavorable

immune response (as it will be discussed) (110, 111). Adaptive immune response is

developed during the lifetime of an individual as an adaption to infections with that

pathogen. Thus, creating a memory of defense against this pathogen in the secondary

infection is an important role of adaptive immunity known as immunological memory

which confers lifelong protective immunity. Other non-structural protein such as NS3 and

NS5 could be also a target of antibodies response but, these antibodies generate a weak

immune response. The possibility of presenting those antigens (NS3, NS5) to B-cells are

limited and depend on the lysis of infected cells to release those antigen into the

extracellular space (3). The main role of vaccination is to generate lifelong protective

immunity against certain pathogens. Protective immunity against secondary infection of

DENV is one of the most important consequences of adaptive immunity which depend on

generating a population of memory lymphocytes which can mediate a long-lived

immunological memory to generate rapid and strong immune response in the secondary

infection of DENV infection. Individuals who have been infected with one dengue

serotype (Primary DENV infection) have long-term protective immunity against that

serotype and short-term protection against the other three serotypes of DENV (3).

Page 50: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

36

Subsequently, the patient will be in danger to a sever disease after the secondary infection

of the other three serotypes, known as immunopathogenesis of DENV infection.

1.5. Immunopathogenesis of Dengue virus secondary infection: Immune response

against DENV infection has two possible outcomes; protection against infection and

illness or unfavorable clinical outcomes such as DHF and DSS . The well-established

finding that there is a high chance of generating severe illness such as DHF and DSS in

patients with secondary infection of heterotypic Dengue serotypes provides strong

evidence for pathological side to DENV pre-existing immunity. Protective immune

responses against one serotype are capable to respond more rapidly than naïve cells

during the secondary infection of other serotypes (98, 112, 113). However, because of

sequence diversity between the DENV serotypes, the re-activated immune response

occurs in the context of serotype cross-reactive immune response. Antibody-dependent

enhancement of dengue infection can be mediated by anti-E protein antibodies, which

trigger the uptake of mature DENV by host cells, and anti-PrM antibodies, which trigger

the uptake of immature or partially mature DENV by host cells. For instance, up on the

secondary infection of other serotypes the memory B and T cells will be reactivated to

fight the infection. But these reactivated lymphocytes do not have the optimal degree of

affinity for the new infecting virus epitopes that result in unfavorable immune response

or immunopathogenicty. The major factors that are considered to contribute to the DENV

immunopathogenicty are Anti- Dengue antibodies (114), serotype cross reactive T cells,

complement activation and soluble factors.

Page 51: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

37

Antibodies responses associated with disease, some evidence suggests that

specific antibodies play a role in the viral pathogenicity (35). Such evidence includes the

increment of the risk to develop DHF and DSS during primary infection in infants whom

born to mothers with established immunity to DENV. The tropism of DENV for

monocytes and dendritic cells, both expressing FcδR receptor for the heavy chain of the

IgG, creates the opportunity for DENV specific antibodies to enhance the viral entry (see

figure 10) (115). A phenomena called “antibody- dependent enhancement” (ADE) it

enhances the disease (116). This phenomenon explains the risk of developing DHF and

DSS occurred in infants (at age of 6 to 9 month) who were infected with a DENV

serotype different from that which infected their mothers. The infants receive anti-dengue

maternal antibodies that declined to sub-neutralizing levels. In that case, the number of

the antibody molecules bound per dengue virions is below the threshold necessary for

neutralization (116). Thus, the host cell covered by FcδR receptor will up take the

virions- antibodies complex more efficient than the entry of the virus through the viral

specific receptors which result in increasing the viral loads and enhancing the disease

(35).

T cell responses associated with disease: activation of the memory T cells with

higher avidity for the primary infection viral serotype than the new infecting serotype

generates “cross reactive T cell response” which case tissue damage as a result of

cytolysis or inflammation induced by the high number of activated memory cells. During

secondary infection with a heterologous serotype, cross reactive epitope reactivate a large

number of memory T cells than naïve cells (117). These cross reactive T cells have low

Page 52: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

38

avidity to the viral epitope serotype of the secondary infection, which will release

cytokines such as TNF-α and IL-13 which affect vascular permeability. Soluble factor

associated with disease, the activation of high numbers of non-protective T cells and the

high viral load or the enhancement of the disease due to ADE, result in a “storm” of

inflammatory cytokines and other mediators leading to plasma leakage. The main

characteristic of DHF/DSS is the plasma leakage that is caused by malfunction of

vascular endothelial cells induced by cytokines or chemical mediators. The cytokines

elevated in patients with DHF/DSS includes, TNF-α, IL-2, IL-12and IL-6. OF those

cytokines TNF-α plays a critical role, there is a positive correlation between TNF-α

concentration and thrombocytopenia as it can induce endothelial cells apoptosis and

plasma leakage (23, 111). Complement activation associated with disease, the increment

of the viral load during the secondary infection results in a high level of secreted NS1

which will be recognized by cross-reactive antibodies; this complex activated the

complement specially C3a and C5a which induce plasma leakage. This evidence

suggested that a balance between the favorable and unfavorable immune response

determines the outcome of DENV infection. Therefore, there is a high urge to generate

vaccine alternatives to provide protection against the DENV infection and the

unfavorable immune response against the virus infection (23).

1.6. Dengue virus vaccine research and development progress: The first attempt to

generate Dengue vaccine was in 1945 by A.Sabin who attempted to generate an

attenuated DENV; the virus was reactogenic caused a rash in human volunteers (9). Till

now there is no licensed Dengue vaccine in the market due to the challenges of Dengue

Page 53: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

39

vaccine development(see table 1). These challenges include the multiple viral serotypes,

incomplete cross- protection, and the development of ADE, theoretical potential for DHF

and DSS in vaccinees. In addition to, viral interference and lacking animal model that are

faithful to human disease. Instead, Dengue vaccine should be free from significant

reactogenicty and induce long life protection against four Dengue serotypes to avoid

enhancement of the disease (3). Currently, there are many dengue vaccine candidates in

the preclinical and different clinical testing phases (see figure 11 and table 1). Vaccine

development has fixed stages of testing the new candidates that includes: Pre-clinical,

Phase I, II and III. Pre-clinical studies which includes animal model experiments. Clinical

testing which divided into three phases; Phase I is the first attempt to test the candidate

vaccine in humans involves a small group of adults (20-80). It aims mainly to verify the

safety in human. Phase II includes larger group of several hundred individuals. It is

divided into two sub stages, Phase IIa and Phase IIb. Phase IIa includes groups at risk of

acquiring the disease and present in the endemic areas. Phase IIb includes groups at low

or no risk of acquiring the disease. The aims of phase II are to study the candidate

vaccine’s safety, immunogenicity, proposed doses, schedule of immunizations, and

method of delivery. Phase III is the last clinical trial and includes larger trials, involving

thousands to tens of thousands of people in different parts of the world. To be approved

and licensed the vaccine should pass a Phase III trial successfully. The vaccine developer

will submit a Biologics License Application to the FDA. Finally, the candidate vaccine

may be license to be manufactured. Ideal Dengue vaccine could be a live attenuated virus

Page 54: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

40

vaccine or chimeric virus vaccine or an inactivated whole virus, protein subunit vaccine,

a vectored vaccine or a DNA vaccine.

Live attenuated vaccine candidates: When passing the virus serial passages in

tissue culture. The virus will lose its virulence or became less virulence that is known as

attenuation. Live attenuated virus is a viable virus that can replicate in host cells but is

less harmful than the wild type virus. Basically, the attenuated virus carries mutations in

its genome that cause the changing in the virulence of the virus and are not present in the

wild type virus. Attenuation can be reached by passaging the virus several passages in

tissue culture or by genetics technique to introduce specific mutations in the viral

genome. Live attenuated DENV vaccine can induce strong humoral and cellular immune

responses to both structural and non-structural proteins as it mimics the natural DENV

infection. Live attenuated virus vaccine is a cost efficient vaccine. It replicates to high

titer in tissue culture to greater than 107 infectious unit per ml in cell culture, which

means that 100 ml cell culture can yield nearly 1 million doses of vaccine, as the

attenuated DENV can infect human at low doses of 103 infectious unit. However, the live

attenuated DENV vaccine has its own challenges. The replication of a live DENV

vaccine can develop a significant illness that generates symptoms such as fever, rash,

headache and pain. The risk of transmissibility by mosquitos should be considered as the

replicated DENV vaccine can generate high titer viraemia, which is sufficient to initiate

an infection in the mosquito. Genetic instability of the mutations that confer the

attenuation phenotype can cause DENV reactogenicity following replication in humans,

which may lead to development of severe illness.

Page 55: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

41

Page 56: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

42

Some of these challenges were solved using the guidelines of Yellow fever live

attenuated vaccine, such as the doses of the vaccine that generates only low titer viremia

which reduce the risk of transmissibility by mosquitos and develop asymptomatic or sub-

clinical sign of infection after vaccination (118). Live attenuated DENVvaccines are

under development; Walter Reed Army Institute of Research (WRAIR) generated

attenuated DENV 1,2,3,4 by passages the virus in primary dog kidney (PDK) cells and

terminal passage in fetal rhesus lung cells. The candidates were passed 20 times in PDK

cells and the attenuated virus monovalent DENV1-2-3-4 (PDK 20) vaccine candidates

have been evaluated in rhesus macaques where it induce neutralizing antibodies to clear

Dengue infection. These monovalent vaccines were tested in phase I trial where DENV2-

3-4 vaccines were mildly reactogenic, but DEN1 retained its reactogenicity and generated

rush and fever in 40% of the vaccines (119). A the result of phase I, DENV-1 was

considered under-attenuated and generated excessive immune response and DENV-4 was

over attenuated and generated poor immune response. Thus, a further passaged DENV-1

was generated by passage the virus in PDK 27 times and a lower passaged DENV-4 was

generated by passage the virus in PDK 6 times instead of 20 times. Currently, tetravalent

formulation composed of DENV-1(PDK27), DENV-3(PDK20), DENV-4(PDK6) and

DENV2 (PDK53) are in phase II trial in 86 adult volunteers in North America and South

Asia. DENV-2 was the only exception that was different in phase II from Phase I. As

DENV-2 was passaged 53 times in PDK cells, the virus was not inducing viremia while

stimulating robust immune response(120, 121). In order for WRAIR vaccine to pass the

clinical trials, the mutation contributing to the attenuation phenotype should be identified

Page 57: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

43

and show how stable they are (122). The National Institute of Allergy and Infectious

Diseases (NIAID) used reverse genetic techniques to introduce defined deletion mutation

in the 3’-untranslated region (UTR) of DENV-1-4 full-length cDNA clones to generated

attenuating DENV-1-4. The deletion of 30 nucleotides at the 3’ UTR (nt 9,834-9,863) in

DENV-1-4 generates balance between the level of attenuation and immunogenicity in

non-human primates and humans but not for DENV2-3. During the phase I trial of

rDENV-1∆30 and rDENV-4∆30, 28 volunteers were vaccinated with 10(3) plaque-

forming units (PFU) of the attenuated virus which induced neutralizing antibodies by 28

days post-vaccination. At day 180, 94% of the vaccinees generated neutralizing

antibodies and generated no symptoms or sub-clinical symptoms. The rest of the

volunteers received a high dose of 10(5) PFU and generated symptoms including rash

and increases in liver enzymes. Phase I trial proved the safety of the vaccine, as it

remained genetically stable following the replication in human, the vireamia titer level

was low and the virus was not transmitted to mosquito because the ∆30 mutation

attenuates the virus replication in mosquito (123). As rDENV-2∆30 and rDENV-3∆30

were not immunogenic in non-human primates, the NIAID used an alternative approach

to generate suitable vaccine candidates for DENV-2-3. Chimeric virus strategy has been

used to introduce the membrane precursor (PrM) and E protein of DENV-2 and DENV-3

into rDENV-4∆30 genetic background to generate the chimeric attenuated DENV-3/4∆30

and DENV-2/4∆30. The two chimeric attenuated viruses and the two attenuated viruses

have been combined to generate a tetravalent DENV vaccine formulation (TetraVax-

DV). TetraVax-DV induce neutralizing antibodies in rhesus monkeys and was used to

Page 58: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

44

immunize volunteers in phase I clinical trial. Acambis and Sanofi Pasteur generated a

chimeric DENV vaccine using the Chimervax platform. Acambis substituted the genes of

the PrM and E protein from each of the four Dengue serotypes into the live attenuated

YF17D vaccine strain (124). The four separated functional chimeric virus were used as

monovalent vaccine candidates and passed the phase I trial. The trail proves that the

monovalent vaccines are safe, as they induce low titer vireamia, generates sub-clinical

symptoms and are noninfectious to mosquitos (124). The four chimeric viruses are then

combined to generate a tetravalent chimeric DENV vaccine, which induce a high level of

neutralizing antibody in monkeys. The phase I testing of ChimeriVax tetravalent vaccine

was safe without any serious adverse side effect and immunogenic. However, a phase IIb

clinical trial showed poor efficiency as the vaccine doses generate protection against

DENV-1-3-4 but not DEN2, which negatively impacted the overall protection.

ChimeriVax is the most advanced vaccine candidate to be tested in humans. The Center

for Disease Control and protection (CDC) in the US used the attenuated DENV-2 PDK-

53 developed by Mahidol University to generate a set of chimeric DENV vaccine

candidates by substituting E protein from DEN-1-3-4 into the live attenuated DENV-2

PDK-53 vaccine strain (125). The preclinical studies of the tetravalent formulation

(DENVvax) were safe and immunogenic in monkeys, which make the DENvax moved to

Phase I clinical trial. The previous success of inactivated tick born encephalitis and

Japanese encephalitis virus vaccine encouraged many groups to use virus inactivated

vaccine strategy to generate DENV vaccine.

Page 59: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

45

Whole virus inactivated Vaccine candidates: Inactivated virus is a nonviable

viral particle that can not replicate in the host cells. The preparation of an inactivated

vaccine includes the propagation of the virus in cells such as Vero cells, concentration of

the virus particles by ultracentrifugation, purification on sucrose gradient, and

inactivation of the virus by chemical treatment such as formalin. The virus-inactivated

vaccine has two advantages over the live attenuated vaccines; it is safe, as it is not

possible for the virus to retrieve its viability and cause reactogenicty in vaccinees. It

induces a balanced antibody response and does not result in infection interference.

Nonetheless, the virus-inactivated vaccine has its own challenges. The vaccine cannot be

replicated in the host cells, which means that a high titer of the vaccine should be

generated in tissue culture, which can be expensive to manufacture as DENV does not

grow to high titer in tissue culture. Moreover, the vaccine contains only the DENV

structure protein and fails to induce immune response to the non-structural proteins.

Adjuvants and multiple booster of doses are required to provide long term immunity.

These challenges make the DENV inactivated vaccine not attractive for manufacture in

endemic area, but they may be used as military vaccines. Walter Reed Army Institute of

Research (WRAIR) used a high titer (about 10(9) pfu per ml) purified Dengue 2 virus to

be inactivated with formalin. The inactivated DENV-2 vaccine was used with adjuvant

such as AS05 and AS08 to immunized macaques, which induce high neutralizing

antibody levels after multiple doses (1).

Page 60: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

46

Recombinant subunit vaccines candidates: DENV antigens, primarily E

protein, have been expressed in several systems to generate protein subunit vaccine

candidates. Escherichia coli bacteria, Pichia pastoris yeast, insect cells and mammalian

cells have been used to generate recombinant Dengue protein such as PrM-E and C

proteins (1). Subunit vaccine has the same advantage and disadvantage of the inactivated

vaccine, however one principle should be considered when this strategy is used to

generate subunit vaccine based on E protein. The E protein contains the major antigenic

epitopes of DENVand the primary used antigen in the production of subunit vaccine;

thus, E protein should be folded correctly to induce the right immune response. Full-

length E protein without co-expression of PrM protein will not be folded correctly, will

be targeted intracellular, not secreted and fail to induce immune response (1). Hawaii

Biotech generated a recombinant subunit DENV2 E protein comprised of the N-terminal

of 80% of E and PrM (r80E) (the E protein doesn’t encode 83 a.a at its C-terminal)

expressed in Drosophilae cells. The preclinical trial includes the immunization of rhesus

macaques at day zero and 30 with two doses of r80E with each of five different adjuvant

combinations. Control groups included animals immunized with saline, purified

inactivated DENV-2 given with adjuvant or a single dose of live attenuated DENV2

vaccine. This induced the same level of neutralizing antibodies as in the group that

received the highest dose of antigen together with two adjuvants (1).

DNA vaccines candidates: Comprising DENV genes into an expression plasmid

may result in DENV DNA vaccine, which affords advantages over conventional vaccines

including: stability for long periods of time, and resistance to extremes of temperature.

Page 61: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

47

This mean it can be transport at room temperature and decrease the likelihood of

reactogenicity and replication interference. Because the proteins produced by DNA

vaccine are translated and processed within the host cell, they are able to induce class I

MHC-dependent immune response. Nevertheless, DNA vaccine has disadvantages

including: the necessity for multiple doses, using adjuvants and specialized injection

equipment. Naval Medical Research Center (NMRC) introduced the DENV-2 PrM gene

and 92% of the E gene (the E protein doesn’t include 54 a.a at its C-terminal) into the

plasmid expression vector VR1 012. Preclinical studies in mice and monkeys carried by

immunizing animal by two doses of the DENV-1 DNA vaccine combined with adjuvant

were followed by challenging the animals with wild type DENV-2. The vaccine protected

80% of the animals where the neutralizing antibody was induced in all vaccinated

animals (126).

Live-virus Vectored Vaccines candidates: Recombinant viruses expressing

foreign proteins can induce strong humoral and cellular responses against infection in

humans. The recombinant virus infects cells and expresses its proteins along with the

foreign proteins within the cells. Those proteins are then naturally processed,

glycosylated and associated with the cell membrane. The intracellular translation and

processing of the proteins can induce class I MHC- dependent immune responses.

However, the live-virus vectored vaccine will induce a restricted immune response

depending on the foreign vectored antigen. The National Institute of Allergy and

Infectious Diseases (NIAID) used the highly attenuated Modified Vaccinia Ankara

(MVA) virus to express DENV-2or DEN-4 PrM and 80% of E protein. Preclinical studies

Page 62: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

48

in mice show that the MVA-DEN2 80% E, but not the MVA-DEN4 80% E, induce

neutralizing antibody responses in mice. Monkeys immunized twice with the MVA-

DEN2 80% E develop resistance to wild type DENV-2 challenging (127). Naval Medical

Research Center (NMRC) generates two bivalent DENV vaccines using complex

adenovirus (CAdV) by introducing the PrM and E genes of DENV-1 and DENV-2

(CAdVaxD1-2) or DENV-3 and DENV-4(CAdVaxD3-4). Preclinical studies include

immunization of rhesus monkeys by tetravalent dengue vaccine formulated by combining

the two bivalent vaccines (CAdVaxD1-2) and (CAdVaxD3-4). The vaccine induced

neutralizing antibody at high levels and protected the challenged animal with wild type

DENV, which was administrated at 4 and 24 weeks post-immunization. CAdVaxD3-4

completely blocked viremia while CAdVaxD1-2 significally reduced viremia in

challenged animals (128). All these current studies building toward realizing efficient

vaccination were faced with many challenges; however, the major challenges are

generating tetravalent dengue vaccine that should induce protective response and prevent

any theoretical risk of developing more serious diseases such as DSS and DHF, in

addition to being safe, stable and coast effective vaccine. In this study we will present a

vaccine candidate that has the ability to overcome these challenges using the live virus

vector strategy.

Page 63: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

49

7

Figure 11. Representation of various Dengue vaccine candidates antigen make up. A.

Shows a schematic of the dengue virus genome. B. Acambis and Sanofi Pasteur vaccine

contains for chimeric live flavivirus. The ChimeriVax tetravalent vaccine derived from the

yellow fever virus genome with the precursor membrane (Pr-M) and envelop (E) protein

gene segments replaced by the gene segments of each of the four serotypes of Dengue

virus. C. The National Institute of Allergy and Infectious Diseases (NIAID) and the US

National Institute of Health (NIH) vaccine contains a mixture of four recombinant dengue

virus genome; DENV-1 and DENV-4 have an attenuation mutation on their 3’

untranslated region (UTR) known as ∆30 that are not shown in the figure. The DENV-2

and DENV-3 component are chimeric dengue viruses derived from DENV-4 mutated

genome with PrM and E genes replaced by serotype 2 and 3 of Dengue virus. Chimeric

DENV-4-3 is not shown in the figure D. The Center for Disease Control and protection

(CDC) vaccine contains a mixture of four recombinant DENV-2 genomes; the DENV-1,

DENV-3 and DENV-4 components are chimeric dengue virus genome. The Hawaii

Biotech vaccine is a recombinant protein vaccine containing the E glycoprotein of the four

serotypes of Dengue virus. Other vaccines using DNA plasmid or virus vectors to express

Dengue virus antigen inside the cell. Nature Reviews Immunology(3)

Page 64: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

50

1.7 Hypothesis and research specific aims: We hypothesize that Measles virus (MV)

backbone can be used as a vaccine vector to generate protective responses against both

measles and DENV. We planned to introduce DENVantigens coding sequences

downstream of N gene in our viral vector MVvac2. The virus vector will replicate

effectively in tissue culture and the expression profile of Dengue antigens will be high.

Since recombination or deletion has not been demonstrated in MV, we propose this

vaccine will be stable enough to be tested in experimental animals for immunogenicity

studies.

1.7.1 Research specific aims:

I. Generating MVvectoring DENV2-4 glycoproteins:

A. Amplification of two forms of DENV PrME; Full length form which will

include the coding sequence of the PrM-E glycoproteins (DENV2 strain 16681, PrM

amino acid sequence (166 a. a.) ,and E is 495 a.a based on reference Hahn et la. DENV4

strain ATCC VR 217 PrM (166 a.a) and E is 493 a.a based on reference Zhao et al.)

which will be translated and processed intracellularly. This protein expression plan has

the potential to induce the formation of virus like particles. Soluble form which will

include the PrM-E proteins without the transmembrane domain of E protein (does not

include 45 a.a at the protein C-terminal, DENV2 sE does not include a.a # 730 to 775,

DENV4 doesn’t include a.a # 728 to 773). The protein will be secreted to extracellular

space by the cellular secretory system.

Page 65: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

51

B. Cloning Dengue antigens into MVvector; each of the two forms of either

DENV2 or DENV4 will be inserted downstream of N gene in the MVvector MVvac2

(see figure 7).

II. Recovery of recombinant MVvector:

A. Using reverse genetic technology, MVvectoring Dengue antigens will be

rescued from a full-length infectious cDNA plasmid in vitro.

B. Replication fitness of recombinant MVwill be characterized by multi-step

growth kinetics in Vero/h SLAM cells.

III. Characterization of measles and Dengue glycoprotein expression

A. Corroboration of insertion of Dengue glycoprotein coding sequences using RT-

PCR. The insertion of Dengue genes will be detected from total RNA obtained from

infected cells.

B. By western blot, a monoclonal antibody directed against Dengue glycoprotein

E will be used to corroborate expression of the vectored antigens. Anti Measles

glycoprotein H or N will be used as controls.

C. Indirect Immunofluorescence assay, polyclonal sera directed against Measles

glycoprotein N (positive control) and monoclonal antibodies against Dengue

glycoproteins PrM and E will be used to corroborate the expression of antigens.

Page 66: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

52

Figure12. Representation of Measles Virus structure, RNA genome and proteins. a,

Measles virion structure and organization. Measles virus is negative single stranded RNA

virus contains a lipid envelop with two viral glycoproteins, MV-H and MV-F bind to Matrix

(M) protein though cytoplasmic tail. The genomic RNA is covered by nucliocapsid (N)

protein and bind to phosphoprotein (P) and Large (L) protein to form RNP complex. b, the

organization of measles RNA genome. It encodes 6 genes. Structural genes includes F,H and

M and non-structural genes includes L and p which includes two overlapping open reading

frames C and V. The 3’ end of the genomic RNA has the leader sequence where the RdRp

will bind to initiate the transcription. RdRp generates a transcription gradient of measles

mRNAs. N protein has the higher expression level.

Page 67: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

53

1.8 Measles virus Vector: Measles virus (MV) is a negative sense – single stranded

RNA virus of the Paramyxoviridae family. The MV proteins Include, Nucleocapsid (N)

protein, Phosphoprotein (P), Hemagglutinin (H), Fusion (F) protein, Matrix (M) protein

and an RNA-dependent RNA polymerase (L) protein (figure 12 a, b) (5). Measles is an

enveloped pleomorphic virus, it has two glycoproteins H and F arranged on the surface of

the virion. H is a type II integral membrane protein that is responsible for attachment of

the virus to a cell receptor present on the cell surface. The MV has 3 known receptors,

which determine the tropism of MV within the host cells. CD 46 (membrane cofactor

protein) present on the surface of almost all nucleated human cells(129). CD 150 or

SLAM (Signaling lymphocyte Activation Molecule) marker on the surface of activated

lymphocytes. (130) Nectin-4 is a tight junction protein that mediate the MVentry into

epithelial cells (131). The other glycoprotein is the F protein. It is a type I integral

membrane protein which present in an immature form called F0 and will be cleaved by

cellular proteases prior to virus releasing from the cells in order to expose its hydrophobic

fusion peptide. Mature virus has two subunit of F protein F1 and F2 that is responsible

for the fusion and the entry of the virus into the host cells. When the virus binds to the

cell surface receptor, F1/F2 protein will go through conformational changes at neutral pH

where the fusion peptide will be inserted into the plasma membrane. The virus membrane

will be fused into the cell membrane and the virus genetic material will be released. Both

H and F proteins are integrated to the virus membrane through cytoplasmic tail, which

are bound to the Matrix (M) protein. M protein lines the inner surfaces of the virus

envelop. It plays a role in the virus assembly and provides a support for the glycoprotein.

Page 68: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

54

After the virus entry the genome will be released into the cytoplasm of the infected cells.

MV genome is covered by the Nucleocapsid (N) protein and associates with L protein

and P (5). The N protein is the most abundant protein among measles proteins and it

plays a critical role in the viral replication, as it will be discussed. L protein is responsible

for the transcription and replication of the RNA genome. In addition to the L protein the

virus carry the P protein since it is an essential polymerase cofactor. The MV P gene

codes for not only the P protein but also for other two proteins: V and C, which are not

strictly required for viral propagation in cultured cells but required for the viral

replication in the host cells (see figure 13). The V and C protein play a role in the control

of the innate immune response, antagonizing the interferon pathway.

Page 69: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

55

Figure 13. Representation of Measles virus replication in the cytoplasm of infected cell.

Negative stranded RNA genome covered by N protein and bind to P and L proteins will be

released into the cytoplasm of infected cells after virus entry. The L protein or the RdRp will

de attach from the RNP complex and will bind to the leader sequence at 3’ end to start the

replication process. The genomic RNA will transcribe into negative strand or antigenomic

RNA which is used as a template for replication and production of more negative stranded

RNA genome. The genomic RNA will be transcribed into viral mRNAs that will be translated

by cellular mechanism into proteins. Measles F and H protein will be directed to the ER to be

glycosylated and secreted to the plasma membrane. The M protein will be bind to the

cytoplasmic face of the plasma membrane. The replicated genomic RNA will bind to the N, P

and L proteins to form RNP complex, which will be transported to the plasma membrane.

Once the RNP bind to the M protein a complex will be formed and the virus will bud from the

cell surface. Current Opinion in Microbiology(5)

Page 70: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

56

The Measles RNA genome is used as a template to be transcribed to generate a

full-length messenger RNA. Thus, the replication of Measles genome happens in two

steps. The first one consist of when the RdRp will bind to the leader sequence at 3’ end of

the genome to generate a positive stranded, full-length RNA called anti-genomic RNA.

The second one happens when the RdRp use the anti-genomic RNA as a template to

generate multiple copies of the genomic RNA (Figure 13) (5). In comparison,

transcription happens in a multi-step process where the RdRp will bind to the leader

sequence to transcribe each of MV cistrons in a gradient manner. Transcripts close to the

genomic 3’ end are more abundant, N protein has the higher expression level while L

protein has the lowest (Figure 14).

Figure 14. Represent the relative transcript levels of the six measles genes. The measles

polymerase generates transcription gradients of the measles genes. The genes downstream the

3’ end will have lower level of expression. N protein has the highest level while L protein has

the lowest level of expression. The measles genome has an intragenic region between each

gene. The polymerase will attenuate for a second after transcribing the gene and then it will

released from the genome to bind again to the leader sequence at the 3’ end of the genome and

start the transcription all over again. As a result the polymerase will generate different amount

of the viral mRNA. Current Topics in Microbiology and Immunology

Page 71: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

57

The MV has two distinctive features, Measles genome is only biologically active

when it is present as RNP complex, which means that the virus should be encapsidated

into the N protein in order to be transcribed and replicated. As a member of

Paramyxovirus, measles has another distinctive feature called rule of six. Measles has a

restriction for the genome replication. The length of the genomic RNA or the anti-

genomic RNA should be a multiple of six nucleotides in order to be encapsidated into the

N protein. As the RdRp elongates the genomic or the anti-genomic RNA the N protein

monomers will bind to units of six-nucleotides (hexamers). Due to those two features the

RNA genomes that are not hexameric will not be packaged into the RNP complex and

will replication capabilities. The viral mRNAs will be translated in the cytoplasm. N, P

and L proteins will be used in the viral replication while the M protein will be directed to

the plasma membrane. The H and F protein will be directed to the endoplasmic reticulum

to be synthesized and transported via the Golgi apparatus to the plasma membrane. As

the proteins are transported to the plasma membrane H and F protein will be glycosylated

and cleaved to generate mature forms. The glycoprotein complex will be assembled on

the cell surface and will bind to the M protein that binds to the cytoplasmic face of the

plasma membrane (figure 13) (5). Encapsidated genome will bind to the M protein and

afterwards the virus will bud from the cell surface. Generally, most of the new

synthesized virus particles are not released into the extracellular space. The newly

accumulated H and F glycoprotein at the surface of the infected cell membrane can result

in fusion between neighboring cells. Syncytia formation is one of the known signs of

measles infection in the culture cells. Syncytia formed due to the fusion of infected cells

Page 72: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

58

results in passing infectious virions between cells without releasing virus particles (5). In

this research, the MV vector” MVvac2” that was generated from Schwarz vaccine strain

is used as backbone to generate Dengue divalent candidates. From a vial of Schwarz

vaccine, Measles antigenomic RNAs were isolated which used to generate full-length

cDNA of measles genome using RT-PCR. The cDNA was used to generate pB (+)

MVvac plasmid. Three mutation within the polymerase gene of pB(+)MVvac was

corrected (132). The corrected plasmid was named pB(+)MVvac2. To facilitate cloning

of foreign coding sequences into pB(+)MVvac2 vector, additional transcription units

(ATUs) were introduced into the vector in many positions. In this research,

pB(+)MVvac2 with an ATU located in position 1710 downstream the N gene (see figure

16) is used as a virus vector to express DENVantigen. The foreign antigen sequence can

be introduced in intragenic regions, which direct the transcription by RdRp. The genomic

location of the ATU determines how much protein will be expressed. ATUs have been

inserted into different locations including downstream P, downstream H and downstream

L and upstream N genes. Attenuated MV vaccine is one of the most stable, safe and

effective human vaccines. Vaccination using measles vaccine is protective for life against

MV infection. This strong and durable protection should be applied in case of MV

expressing antigen of other pathogens. MV vaccine vector has many advantages in the

means of expressing foreign antigen. In comparing top DNA viruses, recombinant MV

vaccine is safe because it replicates in the cytoplasm of the cells. Thus, the danger of

integration into the human genome is excluded.

Page 73: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

59

Figure 15. Rescue methods of the Measles virus and recombinant measles virus using

standard cell line. Simultaneous transfection of cells that support measles replication like

293 cells. Each of the three plasmid carry N, P protein or T7 polymerase. Plasmids have a

cytomegalovirus (CMV) promoter which bind to the cellular RNA polymerase to express the

N , P proteins and T7 polymerase. pEMc-La has the L protein coding sequence primed by

internal ribosome entry site (IRES) to ensure the translation of the L gene into the RdRp.

P(+)MV will transcribed by T7 polymerase into full length antigenomic RNA which serve as

template for the replication and the production of the genomic RNA. The N and P and L

protein will bind to the genomic RNA to form RNP complex. MV genome as RNP will

generate and express the viral mRNAs into viral protein by cellular machinery. The measles

virus will assemble and released from the cell. Current Topics in Microbiology and

Immunology (2)

Page 74: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

60

In addition, the small size of MV in comparison to DNA virus gives MV advantage of

replicating more robustly. In comparing to other RNA viruses measles has high genetic

stability. Also, MV can carry high load of insert with no restriction by its helical capsid

structure in compare to other RNA viruses.

In summary, the MV vaccine is safe and it has been used in for more than five

decades, with a well-established production and distribution networks, replicate in the

cytoplasm and easy to use in reverse genetic technology . So, using the MV vaccine as a

backbone to generate the Dengue vaccine will make the preclinical tests easier. The

industry of the MV vaccine is a well-established industry. Thus, we can get production

lines for dengue vaccine based on the MV vector much easier. This will make the cost of

the vaccine production cheaper than other vaccine candidates.

A reverse genetic system has been established to allow the rescue of MV from

cloned DNA. The first attempt to rescue MV was done by using helper virus. In this

method, the cells will be infected with vaccinia virus expressing T7 RNA polymerase at

high level. The cells then will be transfected by cDNA of measles genome along with

plasmid encoding N,P and L genes under the control of a T7 promoter which will be

recognized by the T7 RNA polymerase to initiate the transcription of those genes. The N,

P and L protein will be used in the replication of the transcribed genome (2).

Page 75: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

61

The helper virus approach is an effective way to rescue viruses but it is not the

most effective strategy to rescue MV since a cumbersome purification step was

necessary. Thus, the rescued virus progenies should be purified by filtration. MV is easily

inactivated by chemical and physical stress during the filtration process (2). Thus, another

system not including the helper virus should be used in case of MV rescue. Simultaneous

plasmids transfection is more efficient that the helper virus system. A cell line will be

transfected by a group of plasmids. Three plasmids carry T7 polymerase gene, N gene

and P gene. The plasmids have a strong promoter, which will be recognized by the

cellular RNA polymerase III such as CMV (Cytomegalovirus) promoter. N and P protein

will be used to initiate the measles genome replication .T7 polymerase will start to

transcribe the other plasmids which carry the L protein and a full-length cDNA of

Figure 16. Represent the Map of pB(+)MVvac2(ATU)N plasmid. The upper part

represent the plasmid containing an MV genome with vaccine-identical coding

sequence. The genes of MV are represented by arrow dark gray boxes. The T7 promoter and T7 terminator are indicated. They flanked the measles genome to

regulate the transcription by the T7 polymerase. HδR. hepatitis delta virus ribozyme.

The additional transcription unit (ATU) cassette was added in 1710 position within the

measles vector, which located at the downstream of N gene in measles genome. The

bottom part represents the nucleotide sequence of the ATU. The conserved termination, intragenic, and initiation nucleotides are underlined. The restriction site

of the multi cloning site are indicated, which are MluI, BsiWI, NurI, SnaBI, BssHII

and AatII.

Page 76: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

62

measles genome (figure 17). Any cell line that supports the MV replication could be used

in this method. Helper cells (293-3-46) express P protein, N protein and T7 polymerase it

was first used by Redacke to rescue MV( See figure 18) (2). It will be explained in the

material and methods section. The progeny of rescue MV will be propagated by

overlaying the rescue cells by cell line that supports measles entry and replication such as

Vero cells expresses SLAM.

The ability of MV to incorporate ATU into their genome and with the presence of

established rescue system. Measles could be used to insert foreign coding sequence such

as marker genes to localize the replication of MVwithin the host cells and organs and

genes from other pathogen to generate multivalent vaccines. MV vector was first used to

generate multivalent vaccine against Hepatitis B virus (HBV). HB surface Ag (HBsAg)

coding sequence was introduced into multiple locations on measles genome. The

recombinant measles successfully expressed the HBsAg. Furthermore, vectored vaccines

remain protective against a MV challenge in a natural susceptible model (non-human

primates) Thus, the hope that recombinant measles expressing an antigen of other

pathogens has a documented substrate in order to generate multivalent vaccines.

With the high demand to generate effective vaccine for virus members of Flavivirdia

such as yellow fever (YF), Japanese encephalitis, Dengue fever and West Nile fever

(WN) Previous reports have documented the use of MV in developing multivalent

vaccine for at least three Flavivirus.

a) MV expressing a secreted form of E glycoprotein (Es) from West Nile virus showed

that protective responses against measles and WNV could be obtained

Page 77: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

63

simultaneously (133). The coding sequence of the WNV Es was inserted downstream

of P gene in measles genome. WNV protective immune responses were documented

in CD46-IFNarko mice from WNV infection. Those mice are not responsive to INF-

α/β and express on of MV receptors. The controlled animal died within 3-4 days after

WNV infection challenge while the immunized group shows no mortality. As

conclusion, the multivalent vaccine protect highly susceptible animal from WNV

infection (133)

b) In an effort to generate a vaccine, Tangy group introduced the DIII of E glycoprotein

coding sequence fused to the ectodomain of M protein coding sequance into

downstream of P gene in measles genome (134).

c) Hepatitis C virus. HCV infection causes a serious health problem worldwide with no

treatment and no vaccine. Roberto Cattaneo group used MV vaccine to express the

two HCV glycoproteins. The coding sequence of E1 and E2 glycoprotein of HCV

were inserted into downstream of P gene. Neutralizing immune responses against

HCV were generated in immunized mice (135).

Page 78: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

64

Chapter 2. MATERIAL AND METHODS

2.1 Cells and Viruses. Vero/hSLAM cells ( African green monkey kidney cells

expressing SLAM receptor on the cell surface) and the helper 293-3-46 cell line were

maintained as monolayers in Dulbecco’s modified Eagle’s medium (DMEM) supplement

with 5/10% fetal calf serum, and 0.5 (Vero/hSLAM) or 1.2 (293-3-46) mg/ml G 418.

BHK21 cells were grown in minimal essential medium (MEM, Gibco-BRL)

supplemented with 10% fetal calf serum (GIBCO-BRL), 2 g/l sodium bicarbonate, 50

U/ml of penicillin and 50 μg/ml of streptomycin. Life attenuated Schwarz MV vaccine

strain, to prepare virus stocks Vero/hSLAM cells were infected at multiplicity of

infection (MOI) of 0.03 and incubated at 37 °C When cytopatic effect (CPE) of 80%

were reached they were collected in Opti-MEM and infectious particles released by two

freeze-thaw cycles. The virus aliquot were titrated and storage at -80 °C. DENV 2

(16681) and DENV 4(ATCC VR 217) to prepare virus stock Vero/hSLAM cells were

infected by MOI of 1 and incubated at 37 °C for 5 days. Virus aliquot made by collecting

the supernatant from infected cell line. The virus aliquots were titrated and storage at –80

°C.

2.2 Plasmids and Plasmids construction. The plasmid pB(+)MVvac2(HBsAg)N

contains an infectious MV cDNA corresponding the antigenome of the Schwarz measles

vaccine strain and has the sequence of Hepatitis B surface antigen (HBsAg) coding

sequence inserted downstream N gene (See Figure 19) pB (+)MVvac2(HBsAg)N was

used to introduce DENV 2 and 4 coding sequences (PrME and PrMEsol) into

downstream N. pJet plasmid containing the corresponding DENV 2 and 4 coding

Page 79: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

65

sequences was used as an intermediate vector. DENV 2 coding sequences were obtained

from PD2/I/C/30P/A plasmid containing cDNA of DENV 2 and is used to isolate the full

length PrME coding sequence and the soluble PrMEsol coding sequence. pR424-D4

plasmid contains cDNA of DENV 4 and is used to isolate the full length PrME coding

sequence and the soluble PrMEsol coding sequence. PD2/I/C/30P/A plasmid and PR424

plasmid were used as templates to amplify DENV 2 PrME/PrMEsol and DEN4

PrME/PrMEsol, respectively by PCR,. RA-prME DEN2 Fwd forward primer was used in

the reaction of amplifying DEN2 PrME and PrME sol ( see table2). RA-prME DEN2

REV2 reverse primer and RA-prME DEN2 sol REV2 reverse primer were used to

amplify the DEN2 PrME and PrMEsol, respectively (see table2).

Table 2.

Primers to amplify dengue antigen coding sequence.

Name Sequence Hybridization

RA-prME DEN2

Fwd

5’TCAGCACGCGTATGTCTGCAGGCATGATCATT3’ DEN2 cDNA C

gene

RA-prME DEN2

REV2

5’ACGATGCGCGCTTAATCGGCCTGCACCATG3’ DEN2 cDNA E

gene

RA-prME DEN2

sol REV2

5’ATCAGGCGCGCTTATTAAACCCCACTGAAGGCAG3’ DEN2 cDNA E

gene

RA-prME DEN4

fwd

5’ACTGTACGCGTATGTCAACGATAACATTGCTGT3’ DEN4 cDNA C

gene

RA-prME DEN4

rev

5’ACGGTGCGCGCCTACTATGCTTGAACTGTGAAGCC3’ DEN4 cDNA E

gene

RA-prME DEN4

sol REV2

5’ACGGTGCGCGCCTACTAGACTCCTCCAAACATGGT3’ DEN4 cDNA E

gene

Page 80: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

66

RA-PrME DEN4 fwd forward primer was used in the reaction of amplifying DENV4

PrME and PrME sol (see table2). RA-prME DEN4 rev reverse primer and RA-prME

DEN4 sol REV2 reverse primer were used to amplify the DENV2 PrME and PrMEsol,

respectively (see table2). The Forward primers were design to add Mlu I recognition site

and start codon sequence into the 5’ end of the inserts, Also the reverse primers were

design to add stop codon and BssH II recognition site sequence into the 3’ end of the

inserts (see table 2 and figure19). PCR reaction was performed as follows 2ug of the

templates and 0.2ul of phusion DNA polymerase were used in a 50 mL PCR reaction

with annealing temperature of 52 °C The amplified Genes were introduce into pJet

plasmid using pJET PCR Cloning Kit (Thermo Scientific) to generate

pJETDEN2(PrME), pJETDEN2(PrMEsol), pJETDEN4(PrME)and

pJETDEN4(PrMEsol). The cloned plasmids were transformed into XL-1 blue competent

cells and grow over night at 37 °C. Colonies were selected and analyzed to be used in a

Mini-prep using QIAGEN Plasmid Mini Kit ( QIAGEN). The cloned pJet plasmids were

sequenced using pJET1.2 Fwd, 23-mer forward primer and PJET1.2 Rev, 24-mer reverse

primer which are included in the pJET PCR cloning kit (see Table 3). The Cloned pJET

plasmids and the pB(+)MVvac2(HBsAg)N plasmid were digested by MluI and BssH II

restriction endonucleases enzymes. The digestion reactions were incubated at 37 °C for 1

hr. and 25 ul of the reactions were used to run on 0.8% agarose gel to isolated the

*Red italic is Mlu I restriction enzyme recognition sequence *Blue italic is BssH II

restriction enzyme recognition sequence **Bold is start/stop coding

sequence***Underlined additional nucleotides to comply with the rule of six.

Page 81: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

67

digested inserts, DEN2 PrME/PrMEsol and DEN4 PrME/PrMEsol and the

pB(+)MVvac2(ATU)N plasmid using QIAquick Gel Extraction Kit (Qiagen). T 4 ligase

is used to ligate the DEN2 PrME/PrMEsol and DEN4 PrME/PrMEsol into the

pB(+)MVvac2(ATU)N, ligation reaction incubated at 14 °C overnight and 5 ul of ligation

reaction are transformed into XL-1 blue competent cells. Plasmid extracted from bacterial

culture by using QIAGEN plasmid Maxi Prep KIT (Qiagen) Four plasmid constract were

generated pB(+)MVvac2DEN2(PrME)N, pB(+)MVvac2DEN2(PrMEsol),

pB(+)MVvac2DEN4(PrME)N and pB(+)MVvac2DEN4(PrMEsol) (see figure19) . The

plasmids construct were analyzed by restriction enzymes digestion and by sequencing

using 1681-FWD, DEN 2 E Fwd1 and DEN2 Fwd2 primers to sequence

pB(+)MVvac2DEN2(PrME)N and pB(+)MVvac2DEN2(PrMEsol). 1681-FWD, DEN4

E Fwd1 and DEN4 E Fwd2 primers to sequence pB(+)MVvac2DEN4(PrME)N and

pB(+)MVvac2DEN4(PrMEsol) (see Table 3).

Table 3

Sequencing primers

Name Sequence Hybridization

1681-FWD 5’ACTAGGTGCGAGAGG3’ MVvac2 nucleotide

#1681

DEN 2 E Fwd1 5’AAGGAAGTACTGTATAGA3’ DEN2 cDNA E gene

DEN2 Fwd2 5’CAGAGTGCAATATGAAGG3’ DEN2 cDNA E gene

DEN4 E Fwd1 5’AGTGGCTCTGTTAAGAAC3’ DEN4 cDNA E gene

DEN4 E Fwd2 5’ACAACAGTGGTGAAAGTC3’ DEN4 cDNA E gene

pJET1.2 Fwd, 23 5’CGACTCACTATAGGGAGAGCGC3’ PJET1.2/blunt vector

PJET1.2 Rev, 24 5’AAGAACATCGATTTTCCATGGCAG3’ PJET1.2/blunt vector

Page 82: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

68

Mutagenesis. Correction of mutation present in DEN4 E protein domain III by

two step PCR extension and overlapping PCR (see figure18). The DEN4 PrME was used

as template and DNA polymerase and mutagenesis primer (see table 4) in a site specific

nucleotide substitution extension PCR to correct the wrong nucleotide. Two products

were isolated and cleaned up using a DNA Colum. The products were used in an

overlapping PCR reaction using phusion DNA polymerase and 52 °C annealing

temperature.

Table 4

Mutagenesis primers

Name Sequence Hybridization

DEN4- mut.FWD1 5’ACTGTACGCGTATGTCAACGATAA3’ DEN4 E gene

DEN4- mut.FWD2 5’ATGTCATACACGATGTGTTCA3’ DEN4 E gene

DEN4- mut.REV1 5’ACGGTGACGTCTACTATGCTTGAACTG3’ DEN4 E gene

DEN4- mut.REV2 5’GAACACATCGTGTATGACAT3’ DEN4 E gene

2.3 Virus Rescue and syncytium Formation. A transfection mix was prepared by 1 ug

of pEMC-La (plasmid encoding MV L protein) mixed with 10 ug of each plasmid

construct pB(+)MVvac2DEN2(PrME)N, pB(+)MVvac2DEN2(PrMEsol)N,

pB(+)MVvac2DEN4(PrME)N , pB(+)MVvac2DEN4(PrMEsol) and

pB(+)MVvac2(GFP)N which encodes green fluorescent protein and was used as positive

control. The plasmids were mixed with CaCl2 and 2X Hepes-Buffered-Saline (2X HBS)

provided by Calcium phosphate precipitation promega kit (Promega). The mixes were

incubated at room temperature for 30 min and then used to transfect helper 293-3-46 cells

stably expressing MV-N, MV-P, and T7 polymerase (see figure18).

Page 83: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

69

Figure 17. Schematic representation of mutagenesis PCR. Stage A include substitution of

the wrong nucleotide with the correct one using mutagenesis primers. DEN4-mut.FWD1 and

DEN4-mut.REV1 (Bark blue arrows) were used in PCR reaction 1 and generated Product-1.

DEN-4mut.FWD2 and DEN4-mut. REV2 (Red arrows) were used in PCR reaction 2 and

generated product-2. Stage B include overlapping PCR to generate the DEN4 E protein

sequence with the corrected nucleotide. In this stage product-1 and product-2 are combined

without primers. The overlapping PCR includes denaturing of product-1 and 2, re-annealing to

form heteroduplexs of both products and extension using DNA polymerase to generate the

final product.

Page 84: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

70

Figure 18. Representation of the measles virus rescue strategy. Measles virus plasmid

vector MV contain T7 promoter and TǾ RNA polymerase terminator flanking the measles

genes. L-encoding plasmid PEMC-La contain internal ribosome entry site (IRES) to

ensure the translation of L gene into RNA polymerase. Both plasmids will transfect helper

cells (293-3-46) which express the nucleoprotein N, phosphoprotein P and T7 polymerase.

In the cytoplasm of the helper cells T7 RNA polymerases will transcript positive, single

stranded RNA from the measles cDNA plasmid. The positive single stranded RNA will be

encapsidated by THE N and P protein, which are expressed by the helper cells. The RNA

polymerases will transcript a negative single stranded RNA, which represents the measles

genome. The measles virus proteins will be translated from the measles RNA genome.

These proteins will be used for virus assembly. The measles virus particles will released

from the cell to infect other cells (2).

Page 85: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

71

The helper 293-3-46 cells were seeded early into six well plates and incubated at

37 °C for 5-6 hr. to rich 50-60 % confluence. Transfected helper 293-3-46 cells were

incubated at 37 °C over night. After incubation time the cells were heat shocked for 3

hours at 42 °C to induce the translation of L proteins (The RNA polymerase). The heat

shocked helper cells were incubated at 37 °C over night to recover. In the next day, each

well of the six well plates were overlaid into Vero/hSLAM cells which were seeded into

100mm plates(p 100) earlier and incubated at 37 °C to rich 50% confluent. The p 100

plate of each virus was incubated at 37 °C and was examined for syncytium formation.

MVvac2 (GFP N plate was examined by inverted microscope with epifluorescence

illumination (EVOS FL.) using the GFP channel to examine the syncytium with green

fluoresce. The other virus constructs; MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol),

MVvac2DEN4(PrME)N and MVvac2DEN4(PrMEsol) were examined for syncytium

formation by inverted microscope (ZEISS) using 4 X and 10 X magnification objective

lenses. Green syncytium were detected one day later after incubation of overlaid cells at

37 °C (see figure). Picking up single syncytium to grow isolated clones depended on the

level of infection ; Single clone syncytia was collected 4 days after incubation of overlaid

cells in p 100 plates of MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol),

MVvac2DEN4(PrME)N and MVvac2DEN4(PrMEsol). Single clone syncytia was

collected by scrubbing it with 1 ul of Opti-MEM. Each one of the collected syncytium

were than used to infect one well seeded with Vero/hSLAM cells in six well plates. The

six well plates were incubated at 37 °C and were examined for cytopatic effect. Two days

after the incubation, single clones were collected from the six well plates of

Page 86: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

72

MVvac2DEN2(PrME)N and MVvac2DEN4(PrME)N. Three and four days after the

incubation, single clones were collected from the six well plates of

MVvac2DEN2(PrMEsol)N and MVvac2DEN4(PrMEsol)N, respectively. The single

clones were collected by scrubbing the cells from each well by 2 ul Opti-MEM and

transfer it into vial labeled as Virus passage zero and storage at –80 °C. To generate virus

stocks of the four virus constructed, the viruses were passaged in cell culture to propagate

the virus until it reach maximum titers. The virus prep of passage zero was released by

two freeze-thaw cycles. ‘Blind’ passages were made by using 25 ul of passage zero virus

to infect p 100 plates seeded with Vero/hSLAM cells with 50 % confluent and incubated

at 32 °C. The pates were examined for cytopatic effect of 70-80 % , where the infected

cells were collected and the virus particles released by two freeze-thaw cycles and saved

as passage one at -80 °C. MVvac2DEN2(PrME)N, MVvac2DEN4(PrME) and

MVvac2DEN2(PrMEsol) passage one were titrated. MVvac2DEN2(PrMEsol) virus was

passaged two more times until it reached decent titer.

2.4 Virus Titration. MV and recombinant MV were titrated by TCID50 (50 % Tissue

Culture Infectious Dose) titration method which is based on the end-point dilution assays.

Vero/hSLAM cells were seeded into 96 well plates (2x104 cells and 0.25 ml of DEMEM

per well). Incubate the 96 well plates at 32 °C for one hour. A serial dilution of the virus

suspension from 1 to 12 were used to infect 8 replicates of Vero/hSLAM cells seeded

wells. The 96 well plates will be incubated at 32 °C with 5% CO2 supply for 3 days. The

TCID50 was then calculated using the Spearman-Karber method (karber, G. 1931).

DENV 2 and 4 were titrated by plaque assay on confluent monolayers of BHK-21 cells

Page 87: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

73

grown in 24-well plates. A serial dilution of virus suspension from 1 to 8 were used to

infect the BHK-21 cells reached 80 to 90% confluence in duplicate. The 48 well plates

will be incubated 4 hours at 32 °C for viral adsorption, the BHK-21 cell monolayers were

overlaid with MEM containing 3% carboximethil-cellulose (Sigma), 0.5% fetal calf

serum and 2 mM L-glutamine. The cultures were incubated at 37°C for 7 days. After

incubation period the cells was fixed with 10% formalin and staining 0.5% naphtol-blue-

black (Sigma) and then counted for plaque formation. The titer of the virus stock was

calculated in plaque forming units per milliliter (PFU/ml).

Figure 19, Experimental design used in this study. (A) Schematic representation of the DEN

V polyprotein cDNA, DEN V PrME and PrMEsol insert. Signal peptide, double-spanning

transmembrane domain, Mlu I recognition site/ start codon sequences and stop codon/ BSSH

II recognition site sequences are indicated by solid black box, striped black box, striped blue

box and solid blue box, respectively. Schematic representation of recombinant MV. Includes

recombinant MV with HBsAg, DEN2PrME, DEN2PrMEsol, DEN4PrME and DEN4PrMEsol

coding sequences inserted downstream N gene. For details about pB(+)MVvac2 vector (see

figure 16). The recombinant pB(+)MVvac2 plasmids carried Dengue 2 and 4 antigens were

rescued and generated four clone D2-2, A.4, 1.6.2 and 2.4.1.

Page 88: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

74

2.5 Virus Growth kinetics. Vero/hSLAM cells were seeded into six well plates (5 × 105

cells and 2ml DMEM per well) were infected by MVvac2, MVvac2DEN2(PrME)N,

MVvac2DEN2(PrMEsol)N and MVvac2DEN4(PrME)N at an MOI of 0.03 . At the

indicated time points (12, 24, 36, 48, 72and 96 hr.) supernatants were cleared by

centrifugation and saved as extracellular virus suspension, cells were scraped in Opti-

MEM (Gibco) as intracellular virus suspension and subjected to freeze-thaw cycles. The

extracellular virus suspension (released) and intracellular virus suspension (cell-

associated) titers were determined by TCID50 titration. A serial dilution of the virus

suspension from 1 to 8 were used to infect duplicate of Vero/hSLAM cells seeded into 96

well plates. The plates will be incubated at 32 °C with 5% CO2 supply for 3 days.

2.6 RNA extraction and RT-PCR. RNA were extracted from virus suspension of MV,

DENV 2 and recombinant MV using Trizol, 0.2 ml of chloroform to 1ml of Trizol

reagent. The homogenized samples were centrifuged by 12000 X g for 15 min at 4 °C.

the upper colorless aqueous phase was mixed with 0.5ml of 100% isopropanol per 1 ml

of Trizol and centrifuged at 12000 X g for 10 min at 4 C to pellet the RNA. The pellet

were air dried for 10 min and resuspended by 50 ul of water. 1 ug of the RNA extraction

was used with Titan one tube Rt-PCR kit and system (Rache) to amplify DENV 2 PrME

coding sequence using RA-prME DEN2 Fwd , RA-prME DEN2 REV2 and RA-prME

DEN2 sol REV2 primers set ( see Table 2).

Page 89: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

75

2.7 Western Blot Assay. Vero/hSLAM cells were seeded into p 100 plates and infected

at multiplicity of infection of (MOI) 0.5 of MV, recombinant MV and DENV 2. 48 hours

after incubation at 32 °C , the cells were lysed by RSB NP-40 0.5%. The protein lysates

from Vero/hSLAM cells were fractionated by SDS-PAGE gel electrophoresis and

transferred to cellulose membranes (Biorad). The blots were probed with a monoclonal

antibody directed against DENV1,2,3,4 E protein at 1:1000 dillution (a mouse IgG2a cat.

# ab9202 from abcam), against actin at 1:1000 dillution (a mouse IgG from SIGMA) and

a polyclonal antibodies directed against MV N protein at 1:5000 dillution ( a rabbit IgG

antibody provided from R. Cattaneo) and Measles H protein cytoplasmic tail at 1:1000

dillution ( a rabbit IgG antibody provided from R. Cattaneo ) [ Cathomen.T.,H.Y.Naim,

1998]. A goat anti-mouse IgG, and a goat anti-rabbit IgG, alkaline phosphatase

conjugated (Thermo Fisher Scientific) used as a secondary antibody at 1:1000 dilution.

Alkaline phosphatase activity was visualized with Western Blue Stabilized substrate for

alkaline phosphate reagent (Promega).

2.8 Indirect Immunofluorescence Microscopy. Vero/hSLAM cells were seeded into 4

well glass Millicell EZ slides (MILLPORE) and infected at MOI of 0.06, 0.1 and 1 with

MV, recombinant MV DENV2 and 4, respectively. After 48 hours of incubation at 32 C,

the cells were fixed with 3% paraformaldehyde and incubated at 32 °C overnight. The

cells were permeabilized with 0.1% Triton X-100 in phosphate- buffer saline (PBS).

Both primary and secondary antibodies dilution were prepared in 0.1% triton X-100 in

10% FBS serum in 1 X PBS. Two Mixes of primary antibodies were used, monoclonal

antibody directed against DENV 1,2,3,4 E protein at 1:50 dilution (a mouse IgG2a cat. #

Page 90: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

76

ab9202 from abcam) with the polyclonal antibodies directed against MV N protein at

1:300 dilution ( a rabbit IgG antibody provided from R. Cattaneo) and monoclonal

antibody directed against DENV 1,2,3,4 PrM protein at 1:50 dilution (a mouse IgG cat. #

ab41473 from abcam) with the polyclonal antibodies directed against MV N protein at

1:300 dilution ( a rabbit IgG antibody provided from R. Cattaneo .The primary

antibodies incubations were carried out overnight at 4 °C. A mix of goat anti-mouse IgG

coupled to Fitc (Green fluorescence), and a goat anti-rabbit IgG coupled to PE (Red

fluorescence) were used as a secondary antibodies with dilution of 1:300 and incubated at

room temperature for 2 hours. The coverslips were mounted on slides by using

flouromount G with DAPI (Electron Microscopy Science). The slides examined and

images were taken by inverted microscope with epifluorescence illumination (EVOS

FL.).

Page 91: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

77

Chapter 3. RESULT

3.1 Generation of four MVvac2 full-length plasmids vectoring Dengue 2 and 4

glycoprotein coding sequences. After amplification by PCR, the DEN2PrME,

DEN2PrMEsol, DEN4PrME and DEN4PrMEsol coding sequence were inserted into the

intermediate plasmid pJET and sequenced ( Figure 21). Sequencing analysis showed a

mutation in the sequence of DenV 4 E protein domain III (see figure20, a). A single

nucleotide mutation resulted in substitution from arginine to threonine (R300T, number

correspond to E amino acid sequence). This mutation was corrected by PCR mutagenesis

(see figure 17 and 20,b). The corrected DEN4 PrME and DEN4 PrMEsol were inserted

A. B.

Figure 20, Dengue 4 E protein DIII mutation. A. The evidence of the R300T

mutation. The sequence histogram of Den4 E protein showed a substitution of

cytosine by guanine, which result in changing the threonine a.a to arginine, which

has different polarity. B. Separation of the mutagenesis PCR reaction in 0.8%

agarose gel, lane 1 and lane 2 represent the product1 and product2 of the

extension PCR. Lane 3 represent the product of the overlapping PCR with is about

2Kbp.

Page 92: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

78

into pJET and sequenced. Analyzing of these plasmids showed that the mutation was

corrected, the rest of inserts sequence was correct. Then, DEN2PrME, DEN2PrMEsol,

DEN4PrME and DEN4PrMEsol coding sequences were inserted into pB(+)MVvac2

downstream the N gene. Four construct were produced and were analyzed by restriction

endonuclease digestion (see figure 22 and 23). The recombinant MVs plasmids were

double digested with MluI and AatII enzymes, electrophoresis analysis showed two

bands with approximate molecular size of 20 (for the backbone plasmid) and 2.1 and 2

kbp representing PrME or PrMEsol inserts, respectively. Selected clones sequence were

analyzed and showed no mutations in the inserts sequences.

Figure 21, Represent the map of Dengue virus insert. The full length insert includes 12 a.a

of Capsid protein C terminal which represent the signal sequence to direct the proteins into the

ER. full length Premature membrane (PreM). Full length envelope (E) Protein including the

steam and transmembrane domain. The soluble insert sequence are similar to the full length

except for the E protein sequence. The E protein of the soluble insert is lacking 40 a.a at the C

terminal of the E protein which represent the transmembrane (TM) domain. For clarity, the

amino acid sequence represent the Dengue 2 virus amino acid sequences.

Page 93: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

79

3.2 Rescue of four Vectored MVs expressing Dengue 2 and Dengue 4 glycoprotein.

The four plasmid constructs were used to generate recombinant measles virus derived

from a vaccine strains. Measles virus rescue system details are in the material and

methods chapter. MVvac2DEN2(PrME)N and MVvac2DEN4PrME)N viruses were

rescued and amplified to obtain viral stocks with a titer of 107TCID50/ml, after one

passage in the tissue culture. The MVvac2DEN2(PrMEsol)N virus was rescued

Figure 22, Restriction endonuclease screening of recombinant MVs plasmid vectoring

Dengue 2 coding sequence. A, agarose gel separation of digested recombinant measles

vectoring DEN2PrME insert downstream N gene by Hind III, lane 1 through 6 are

experimental lanes. Lane 7 is a negative control. B, selected clones from lane 2 and 6 in A

were analyzed by Mlu I and AatII double digestion. Lane 1 and 2 were experimental

lanesl. Lane 3 was negative control. C, agarose gel separation of digested recombinant

measles plasmid vectoring DEN2(PrMEsol) insert downstream N gene by Mlu I and Aat

II enzymes. Lane 1 and 2 were negative control, lane 3 through 6 were experimental lanes.

Lane 2 and 3 showed positive result.

Page 94: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

80

with a titer of 106 TCID50/ml after three passages in the tissue culture. The

MVvac2DEN4(PrMEsol)N virus was rescued with a titer of around 104 TCID50/ml after

three passages in tissue culture, the virus aliquots were stored at -80 C and were not go

through further experiments on this study.

Figure 23. Restriction endonuclease screening of recombinant MVs plasmid vectoring

Dengue 4 coding sequence. A, agarose gel separation of digested recombinant measles

vectoring DEN4PrME insert downstream N gene by Mlu I and AatII enzymes. Lane 1

through 8 are experimental lanes. Lane 9 and 10 was negative control. Lane 1, 3, 4, 5, 6

and 7 show a positive result. B, agarose gel separation of digested recombinant measles

plasmid vectoring DEN4 (PrMEsol) insert downstream N gene by Mlu I and Aat II

enzymes. Lane 1 through 4 are experimental lanes. Lane 5 and 6 are negative control.

Lane 1, 2, 3 and 4 showed positive result.

Page 95: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

81

To assess the replication efficiency of these vectored MVs ,a multi-step growth

kinetic analysis was performed (cells were infected by MOI 0.03). As shown in figure 24,

the maximum titers reached by MVvac2DEN2(PrME)N and MVvac2Den4(PrME)N

were very close to those of the parental strain, MVvac2 (8 X 106 TCID50/mL and 107

TCID50/mL, respectively 48 hr after infection). Thus, the replication efficiency of these 2

viruses was equivalent to the parental strain. In contrast to, the

MVvac2DEN2(PrMEsol)N has a lower replication efficiency than the parental strain.

Peak titer of about 105 TCID50/ml were reached 48 hours postinfection for extracellular

and of about 8 X 103 TCID50/ml were reached 48 hour postinfection for intracellular

MVvac2DEN2(PrMEsol)N.

Page 96: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

82

Figure 24, Growth kinetic of cell-associated (Intracellular) and cell-free (extracellular)

virus production in Vero/hSLAm cells. A, time course of intracellular virus production in

Vero/hSLAm cells infected with parental strain virus (MVvac2), recombinant MVs:

MVvac2DEN2(PrME)N (D2-2), MVvac2DEN2(PrMEsol)N (A.4) and MVvac2DEN4(PrME)N

(1.6.2). B, time course of extracellular virus production in Vero/hSLAm cells infected with

parental strain virus (MVvac2), recombinant MVs: MVvac2DEN2(PrME)N (D2-2),

MVvac2DEN2(PrMEsol)N (A.4) and MVvac2DEN4(PrME)N (1.6.2). The viral titers, indicated

on the vertical axis were measured 12, 24, 36, 48, 72 or 96 hrs. postinfection for both graph A

and B. For clarity, Averages and standard deviations of three independent experiments are

indicated in this figure.

Page 97: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

83

Figure 25, Detection of Dengue 2 PrME and PrMEsol mRNA expression in MVs,

Dengue2 virus and two recombinant MVs infected cells. The upper panel represent the

separation of RT-PCR products on 1.2 % agarose gel. Lane1 is the PCR negative control,

lane2 is MVs PCR product, lane3 is Dengue 2 PCR product, lane4 is D2-2 PCR product and

lane4 is the A.4 PCR product. Dengue 2 and D2-2 product are at the same molecular size,

while A.4 product is lower that the Dengue 2 and D2-2 products size. The lower panel

represent the separation of total RNA extracted from cells infected with MVs, Dengue2, D2-2

and A.4 viruses. RNA extracted from cells infected with MVvac2 present in lane 1, Dengue2

virus present in lane2, D2-2 virus present in lane 3 and A.4 virus present in lane4. Lane 2

show no RNA, because Dengue 2 virus was collected from the supernatant of the infected

cells. The subunit of the cellular ribosomal RNA can be detected at lane1, 3 and 4.

Page 98: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

84

3.3 Determination of Dengue 2 and 4 glycoprotein expression by vectored MVs. The

expression of Dengue2 and Dengue 4 PrME glycoprotein was detected at the level of

mRNA using RT-PCR and/or at the level of proteins using Western blot and indirect

immunofluorescenc-e assay. For RT -PCR analysis, total RNA was obtained from

infected cells at 48 h post infection. The same primers that were used to amplify this

sequence from full-length plasmids were used in a single-step RT-PCR reaction. As

shown in figure 25, The separation of the RT-PCR products in 1.2% agarose gel showed

that RNA extraction from Dengue 2 infected supernatant, MVvac2DEN2 (PrME)N and

MVvac2DEN2(PrMEsol)N infected cells have amplicons corresponding to the PrME

mRNA. As expected, the amplified band of the MVvac2DEN2 (PrMEsol) N has a lower

molecular size than the one obtained from Dengue2 and MVvac2DEN2 (PrME) N. The

difference in molecular size results from the deletion of the E protein transmembrane and

cytoplasmic domains in case of the PrMEsol insert. Total RNA obtained from infected

Figure 26, Western blot of protein extract by lysis of Vero h-Slam cells non-

infected, infected with MVvac2 , D2-2 , A.4 or Dengue 2 virus. The upper panel

represenst the detection of Measles H protein using anti-H antibodies. The lower

panel represents the detection of Actin protein using anti-actin antibodies. As

shown lane 1 is non-infected cells protein extract, lane 2 is MVvac2 infected

cells protein extract, lane 3 is D2-2 infected cells protein extract, lane 4 is A.4

infected cells protein extract, lane 5 is Dengue 2 virus infected cells protein

extract.

Page 99: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

85

cells showed an expected electrophoresis migration pattern, with the ribosomal RNA

bands evident (figure 25, lower panel). The Dengue 2 and 4 PrME glycoproteins

expressed from vectored MVs was then characterized by western blotting. Protein

extracts of infected cells were blotted into nitrocellulose membrane and incubated with

anti DEN1,2,3,4 E mouse monoclonal antibody to detect the expression of Dengue

glycoproteins. As shown in figure27, anti-E antibodies recognize the E glycoprotein with

molecular mass of 59 KDa (53KDa the mass of the protein and 6 KDa the mass for two

glycosylation sites). As expected the expression of E protein was detected in the extract

of Dengue2, MVvac2DEN2 (PrME) N and MVvac2DEN2 (PrMEsol) viruses infected

cells but not in the extract of MVvac2 infected cells. However, we were unable to

determine the expression of DEN4 E protein on this assay. MVs H and N glycoproteins

were detected using anti H mouse or anti N rabbit polyclonal antibody to detect the

expression of measles proteins. As shown in figure 26 and 27, Anti N recognize the N

glycoprotein with molecular mass of 62 KDa and the anti H recognize the H glycoprotein

with molecular mass of 80 kDa in cells extract infected with MVvac2,

MVvac2DEN2(PrME)N and MVvac2DEN2(PrMEsol)N viruses but not in the Dengue 2

virus infected cells extraction. Actin detection was done using anti-actin mouse

monoclonal antibody as a control of the protein concentration in different cell extract (see

figure 26 and27). Indirect immunofluorescence assay was used to detect the expression of

Dengue 2 and Dengue 4 PrM and E glycoproteins in Vero/hSLAM cells infected with

MVvac2, Dengue2, Dengue 4, MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol)N and

MVvac2DEN4(PrME)N viruses. As shown in figures 29 and 30, the expression of

Page 100: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

86

Dengue2 and 4 E proteins were determined in cells infected with recombinant MVs:

MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol)N and MVvac2DEN4(PrMEsol)N and

in cells infected with Dengue 2 and 4 viruses. The expression of Dengue2 and 4 PrM

glycoprotein was confirmed in cells infected by Dengue 2 and 4 as well as recombinant

MVs: MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol)N and

MVvac2DEN4(PrMEsol)N as shown in figure 28and 31.

Figure 27, Western blot of protein extract by lysis of Vero h-Slam cells

non-infected, infected with MVvac2 , D2-2 , A.4 or Dengue 2 virus. The upper panel

represent the detection of Dengue E protein using anti-E antibodies. The medial

panel represent the detection of Measles N protein using anti-N antibodies. The

lower panel represent the detection of Actin protein using anti-actin antibodies.

As shown lane 1 is MVvac2 infected cells protein extract, lane 2 is Dengue 2 virus

infected cell protein extract lane 3 is D2-2 infected cells protein extract, lane 4 is

A.4 infected cells protein extract, lane 5 is 1.6.2 virus infected cells protein extract.

Page 101: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

87

The syncytium obtained from cells infected by MVs and recombinant MVs, were

screened for the expression of MV N glycoprotein. As shown in figures 28, 29, 30 and31,

the expression of N glycoprotein was detected in all MV and recombinant MVs infected

cells and not in Dengue2 and 4 infected cells. Non-infected cells showed negative results

for anti-E, anti-PrM and anti-N screening. Recombinant MVs: MVvac2DEN2(PrME)N,

MVvac2DEN2(PrMEsol)N and MVvac2DEN4(PrMEsol)N infected cells showed

positive result for the dual expression of MVs N glycoprotein and Dengue virus E or PrM

glycoprotein as shown in figures 28,29, 30 and 31. The cell’s nucleuses were stained blue

by DAPI to visualize the cell nuclei.

Page 102: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

88

Figure 28. Expression of Dengue2 PrM protein from recombinant MVs. Vero/hSLAM

cells were non-infected (A) or infected with either Dengue2(B), MVvac2(C) D2-2(D) or A.4

(E) viruses and processed for indirect immunofluorescence as described in Materials and

Methods. MVs N was detected with rabbit anti-N antibody and Dengue 2 PrM was detected

with mouse anti-PrM antibody, followed by anti-rabbit- and anti-mouse antibody– PE(red)

/FITC(green) conjugate, respectively. The duel expression of N and PrM protein was detected

by overlapping the GFP and RFP channels, as shown in marge Colum. The fixed cells were

stained by DAPI blue, which gave the nuclei this blue color when using DAPI blue channel in

the microscope.

Non-Inf

Degue 2

MVvac2

D2.2

A.4

Page 103: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

89

Figure 29, Expression of Dengue2 E protein from recombinant MVs. Vero/hSLAM

cells were non-infected (A) or infected with either Dengue2 (B), MVs(C), D2-2(D) or A.4

(E) viruses and processed for indirect immunofluorescence as described in Materials and

Methods. MVs N was detected with rabbit anti-N antibody and Dengue 2 E was detected

with mouse anti-E antibody, followed by anti-rabbit- and anti-mouse antibody– PE(red)

/FITC(green) conjugate, respectively. The duel expression of N and E protein was detected

by overlapping the GFP and RFP channels, as shown in marge Colum. The fixed cells were

stained by DAPI blue, which gave the nuclei this blue color when using DAPI blue channel

in the microscope.

Non-Inf

Degue 2

MVvac2

D2.2

A.4

Page 104: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

90

Figure 30, Expression of Dengue4 E protein from recombinant MVs. Vero/hSLAM cells

were non-infected (A)or infected with either Dengue 4(B), MVvac2(C), or 1.6.2(D) viruses and

processed for indirect immunofluorescence as described in Materials and Methods. MVs N was

detected with rabbit anti-N antibody and Dengue4 E was detected with mouse anti-E antibody,

followed by anti-rabbit- and anti-mouse antibody– PE (red) /FITC(green) conjugate,

respectively. The duel expression of N and E protein was detected by overlapping the GFP and

RFP channels, as shown in marge Colum. The fixed cells were stained by DAPI blue which gave

the nuclei this blue color when using DAPI blue channel in the microscope.

Non-Inf

Degue 4

MVvac2

1.6.2

Page 105: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

91

Non-Inf

Degue 4

MVvac2

1.6.2

Figure 31, Expression of Dengue 4 PrM protein from recombinant MVs. Vero/hSLAM

cells were non-infected (A)or infected with either Dengue 4 (B), MVvac2 (C), or 1.6.2(D)

viruses and processed for indirect immunofluorescence as described in Materials and

Methods. MVs N was detected with rabbit anti-N antibody and Dengue4 PrM was detected

with mouse anti-PrM antibody, followed by anti-rabbit- and anti-mouse antibody– PE(red)

/FITC(green) conjugate, respectively. The duel expression of N and PrM protein was

detected by overlapping the GFP and RFP channels, as shown in marge Colum. The fixed

cells were stained by DAPI blue, which gave the nuclei this blue color when using DAPI

blue channel in the microscope.

Page 106: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

92

Chapter 4. DISCUSSION

Dengue infections are significantly affecting South -East Asia (SEA), the Western

Pacific, and in some countries in South Africa. “Of the total world population of 6.2

billion, countries of the South-East Asia Region (SEAR) account for 1.5 billion (24%).

On that scale, of the 2.5 billion people (living in the tropics and sub-tropics) at risk of

DF/DHF, 52%, i.e. 1.3 billion populations, live in SEAR” (136). These countries include

Thailand, India and Indonesia. Dengue also causes major health problems in the Western

pacific countries, including Malaysia, Philippines and Vietnam. In South America,

countries, such as Mexico and Peru, dengue infections are the cause of hospitalization

and death among children. Preventing Dengue infection is a public health priority.

Because there is no specific treatment for the disease and the control methods such as

chemical sprays and windows screams fail to efficiently limit the transmission of the

virus, there is a priority to develop an efficient vaccine. Major challenges for this

development are related to market- and technology- mainly. The market issues include

vaccine pricing and demand projections. Since, the most affected countries are

developing countries, there is a need to generate a cost-efficient vaccine. In addition, the

geographical expanding of dengue virus infection increases the concerns about vaccine

demand projection. Technology related issues include the safety and the need to produce

simultaneous protective immune responses to all four serotypes. Candidate vaccines that

use diverse approaches are currently in various stages of development (table one) are

facing difficulties immunizing epidemic areas. Live attenuated vaccines including

attenuated viruses, recombinant viruses and chimeric live attenuated virus vaccine

Page 107: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

93

approach face viral interference and recombination. For instance, the chimeriVax

tetravalent Dengue vaccine candidate generated by Acambis and Sanofi Pasture is facing

the challenge of viral replication interference. When monovalent candidates are tested,

positive result are encouraging, however, a tetravalent formulation tested in human,

higher doses are required to induce a high level of neutralizing antibodies against the four

Dengue serotypes (124). The viral replication interference happen because the four live

attenuated viruses are competing to enter the host cell through the same cell receptor

which result in decreasing the replication profile of the live attenuated vaccine

candidates. Recombination is another concern when using a tetravalent live attenuated

virus vaccine. The RNA viruses can naturally recombine within and between species.

One example is the recombination between a Sindbis-like virus and the eastern equine

encephalitis virus that resulted in emergence of Western equine encephalitis virus (137).

The homologous recombination among flavivirus has been reported by Seligman. S. J., et

la in 2004, which introduce concerns about the tetravalent live attenuated dengue vaccine

safety(138). In contrast, inactivated vaccines face the problem of cost efficiency since a

high dose of purified and inactivated virus introduce along with adjuvant makes the cost

of producing this alternative very high. Subunit vaccines share the same challenge. The

purification process and the need for adjuvants to induce high neutralizing antibody titer,

increase the cost of the subunit vaccine development. Both inactivated and recombinant

subunit dengue vaccine candidates are not acceptable vaccine for endemic area.

Page 108: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

94

Plasmidic DNA expressing Den 2 glycoprotein face cost and safety challenges as well as

poor antigenic presentation. Kochel et al. at NIMRC try to enhance the trafficking and the

presentation of the den 2 antigen by fusing the protein to a lysosome associated

membrane protein which enhance the immunogenicity of the vaccine, but it is still

considered of low immunogenicity (126).

In this research, the virus vector MVvac2 generated from Schwarz vaccine strain

is used as a backbone to generated a Dengue vaccine. We propose that MV- DenV

divalent vaccine will overcome all the challenges faced that was mentioned for the DenV

vaccine candidates. The recombinant MVs was experimentally tested for genetic stability,

safety and efficacy. It showed genetic stability which is unexpected for RNA viruses, and

ability to carry a high load of genetic material. In addition to the low production cost per

vaccination dose (review the introduction section for more details). All this factors will

make MV- DENV divalent vaccine a suitable vaccine candidates to be used in endemic

countries. However, we are aware that the preimmunity to measles virus could be a

concern if the vaccine will be used in adolescent and adults. In animal models, the

preimmunity from vaccination with MV vaccine does not prevent a boost with

recombinant MV. In fact, boosts enhance the immune response against MV and the

transgene(139, 140). For instance, measles immunity can be boosted in children who are

preimmune after a first vaccination during infancy. In the present study, two different

forms of either DEN2 or DEN4 were introduced downstream MV N gene that resulted in

four different recombinant MVs vector. These virus vectors were used to generate

Page 109: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

95

recombinant MVs. The aim of generating membrane bound and secreted DenV E

glycoprotein is to test which form would induce a higher immune response in animal

models. The recombinant MVs expressing membrane bound (full length) E protein will

replicate in the cytoplasm of the host cells with the potential of displaying DenV E

glycoprotein on the surface of the infected cells. As a consequence, the infected cells will

be a target for CD8+ T-cells and will generate a cellular immune response against Den V

infection. The recombinant MVs expressing secreted (soluble) E protein will replicate in

the cytoplasm of the infected cells and will result in secretion of DenV E glycoprotein to

the extracellular space. As a consequence, neutralizing antibodies directed to Den V E

protein will be secreted from B-cells and will generate a humoral immune response

against Den V infection. The results of this study showed that (i) recombinant MVs

carrying Den V glycoproteins coding sequence downstream N gene can be rescued and

propagated in tissue culture, (ii) the replication profile of the recombinant MVs was

similar to that of the parental strain in case of recombinant MVs expressing full length E

protein but not soluble E, (iii) the two forms of E protein were successful expressed by

the recombinant MVs. The four amplified coding sequence of Den2V PrME, Den2V

PrMEsol, Den4V PrME and Den4V PrMEsol were analyzed to identify any mutation in

the sequence. Both Den4V PrME and Den4V PrMEsol sequence had a nucleotide

substitution mutation at the E protein domain III which would affect the immune

response against DenV infection. The R300T mutation (figure20) resulted in substitution

of threonine that is polar uncharged amino acid by arginine, which is a positive charged

amino acid. We corrected this mutation. The replication profile of the recombinant MVs

Page 110: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

96

varied depending on the expressed Den Ag. As showed in the figure 26, the replication

profile of recombinant MVs expressing Den2V PrMEsol Ag was lower than the

replication profile of the parental strain and the recombinant MVs expressing either Den2

or Den4 full-length antigens. This observation in the replication profile of MVvac2DEN2

(PrMEsol)N can have two explanations. First, we think that the secreted E glycoproteins

were accumulated in the SlAM receptor in the surface of the nearby cells, which may

interfere with the entry of newly synthesized recombinant MV. Which is using SLAM

receptor to enter the nearby cells. Second, we think that the secreted E glycoprotein can

bind to the MV glycoprotein in the secretory exosome. Which interface with the ability of

the recombinant MV to enter the nearby cells. However, we have now demonstrated that

MVvac2DEN2(PrME)N, MVvac2DEN2(PrMEsol)N and MVvac2DEN4(PrME)N are

expressing DenV PrM and E glycoprotein. As shown in figures 28, 29, 30, and 31. The

expression of both Den2 and Den4 PrME glycoprotein were detected by using

monoclonal Ab against either E or PrM glycoprotein in the indirect immunofluorescence

assay. The difference in the molecular mass between the PrME and the PrME soluble was

demonstrated by RT-PCR as shown in figure#. Western blot assay was performed to

detect MV N and H glycoprotein as well as Den2 E glycoprotein. However, the detection

of E glycoprotein in western blot assay was weak. To address these issue we used indirect

immunofluorescence assay to demonstrate the expression of Den2 and Den4 glycoprotein

by recombinant MVs. In conclusion, we have proof that recombinant MVs expressing

DenV glycoprotein have a good replication and expression profile and these recombinant

MVs can be used in future experiments. Future prospective experiments will include:

Page 111: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

97

endoglycosidase digestion experiments with the goal of corroborating the quality of the

antigens vectored by measles; the biophysical analysis of secreted material to

demonstrate the presence of empty viral-like particles containing dengue antigens, and

localization of the expressed full length and soluble E glycoprotein using protein tagging

methods. These experiments can be done for only recombinant MV expressing Den2V

antigen as a proof of concept Immunogenicity study can be done by immunization of

mice susceptible to MV with the recombinant MVs expressing either Den2V or Den4V

antigens. Nevertheless, the experimental results described in this thesis give substrate for

animla experimentation. The first step is the demonstration that our vectors can elicit

neutralizing immune responses against the corresponding dengue virus while still being

able to elicit immune responses against the vector backbone that can be considered

protective. The more suitable animal model for this experiment is our measles-susceptible

genetically modified mouse model. This host expresses one of MV cell receptors (CD46)

with a human-like distribution in a type I interferon defective background 9which allows

for MV replication). This small host (GeCD46-INFrako mice) is a gold standard to discern

vaccine candidates with good in vivo replication and hence advantageous vaccine

potential. During a second phase of animal experimentation, protective responses against

the relevant dengue virus will be assessed in AG129 transgenic mice passively

immunized with reactive sera obtained from GeCD46-INFrako mice. AG129 mice are

defective in type I and type II interferon responses and can replicate mouse-adapted

dengue viruses, but most importantly, this small model reproduces vascular changes upon

dengue infection, hence vaccine responses can be rated in this model.

Page 112: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

98

MVs vector could be modified to generate more robust immune responses, if

needed. These could br achieved by enhancing the fusion ability of MV, which may

increase the spread, and the replication of the virus in Human. Shumpei Watanbe et la,

demonstrated that certain mutation in the Fusion proteins enhanced the fusion activity

and promotes the spread of MV in human. In this study Shumpei demonstrated that

recombinant MVs with mutation in the ectodomain of F protein have enhanced fusion

ability. These viruses can induce cell fusion in SLAM and nectin 4-negative cells unlike

parental strain (141). In addition Shumpei demonstrated that mutation in M protein can

induce the fusion activity of wild type MV F protein. These result suggest that, the

enhancement of MV fusion would increase the virus replication profile. Because the virus

will be able to replicate on cells that express or do not SLAM and nectin-4 receptor in it

surface. The recombinant MV expressing DenV antigen can be modified in the same way

to enhance the virus fusion activity which will result in improving the replication ability

of the viruses. The fusion activity of the F protein can be enhanced by deletion the

cytoplasmic tail of the F protein which interacts with the M protein. Instead of deleting M

protein which may affect the virus particles formation, the deletion of the cytoplasmic tail

of the F protein would prevent the interaction of the F protein and enhance the fusion

ability of The MV. We hypothesize these modified viruses would generate more robust

immune response against both MVs and DenV.

Page 113: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

99

Reference List

1. Whitehead SS, Blaney JE, Durbin AP, Murphy BR. Prospects for a dengue virus vaccine.

Nat Rev Microbiol. 2007;5(7):518-28.

2. Radecke F, Spielhofer P, Schneider H, Kaelin K, Huber M, Dotsch C, et al. Rescue of

measles viruses from cloned DNA. Embo J. 1995;14(23):5773-84. PMCID: 394696.

3. Rothman AL. Immunity to dengue virus: a tale of original antigenic sin and tropical

cytokine storms. Nat Rev Immunol. 2011;11(8):532-43.

4. Perera R, Kuhn RJ. Structural proteomics of dengue virus. Curr Opin Microbiol.

2008;11(4):369-77. PMCID: 2581888.

5. Delpeut S, Noyce RS, Siu RW, Richardson CD. Host factors and measles virus

replication. Curr Opin Virol. 2012;2(6):773-83.

6. Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health, social and

economic problem in the 21st century. Trends Microbiol. 2002;10(2):100-3.

7. Guzman MG, Kouri G, Valdes L, Bravo J, Alvarez M, Vazques S, et al. Epidemiologic

studies on Dengue in Santiago de Cuba, 1997. Am J Epidemiol. 2000;152(9):793-9; discussion

804.

8. Endy TP, Chunsuttiwat S, Nisalak A, Libraty DH, Green S, Rothman AL, et al.

Epidemiology of inapparent and symptomatic acute dengue virus infection: a prospective study of

primary school children in Kamphaeng Phet, Thailand. Am J Epidemiol. 2002;156(1):40-51.

9. Sabin AB. Research on dengue during World War II. Am J Trop Med Hyg. 1952;1(1):30-

50.

10. Souza LJ, Alves JG, Nogueira RM, Gicovate Neto C, Bastos DA, Siqueira EW, et al.

Aminotransferase changes and acute hepatitis in patients with dengue fever: analysis of 1,585

cases. Braz J Infect Dis. 2004;8(2):156-63.

11. Kalayanarooj S, Vaughn DW, Nimmannitya S, Green S, Suntayakorn S, Kunentrasai N,

et al. Early clinical and laboratory indicators of acute dengue illness. J Infect Dis.

1997;176(2):313-21.

12. Kuo CH, Tai DI, Chang-Chien CS, Lan CK, Chiou SS, Liaw YF. Liver biochemical tests

and dengue fever. Am J Trop Med Hyg. 1992;47(3):265-70.

13. Gubler DJ. Dengue and dengue hemorrhagic fever. Clin Microbiol Rev. 1998;11(3):480-

96. PMCID: 88892.

14. Carlos CC, Oishi K, Cinco MT, Mapua CA, Inoue S, Cruz DJ, et al. Comparison of

clinical features and hematologic abnormalities between dengue fever and dengue hemorrhagic

fever among children in the Philippines. Am J Trop Med Hyg. 2005;73(2):435-40.

Page 114: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

100

15. Nimmannitya S. Clinical spectrum and management of dengue haemorrhagic fever.

Southeast Asian J Trop Med Public Health. 1987;18(3):392-7.

16. Butthep P, Chunhakan S, Tangnararatchakit K, Yoksan S, Pattanapanyasat K,

Chuansumrit A. Elevated soluble thrombomodulin in the febrile stage related to patients at risk

for dengue shock syndrome. Pediatr Infect Dis J. 2006;25(10):894-7.

17. Mackenzie JS, Gubler DJ, Petersen LR. Emerging flaviviruses: the spread and resurgence

of Japanese encephalitis, West Nile and dengue viruses. Nat Med. 2004;10(12 Suppl):S98-109.

18. Hardy J. Susceptibility of vector Mosquitoes. In: Monath T, editor. The arboviruses:

Epidemiology and ecology. Boca Raton, FLA: CRC Press; 1988. p. 87-126.

19. Wolfe ND, Kilbourn AM, Karesh WB, Rahman HA, Bosi EJ, Cropp BC, et al. Sylvatic

transmission of arboviruses among Bornean orangutans. Am J Trop Med Hyg. 2001;64(5-6):310-

6.

20. Diallo M, Ba Y, Sall AA, Diop OM, Ndione JA, Mondo M, et al. Amplification of the

sylvatic cycle of dengue virus type 2, Senegal, 1999-2000: entomologic findings and

epidemiologic considerations. Emerg Infect Dis. 2003;9(3):362-7. PMCID: 2958533.

21. Lin CF, Lei HY, Liu CC, Liu HS, Yeh TM, Wang ST, et al. Generation of IgM anti-

platelet autoantibody in dengue patients. J Med Virol. 2001;63(2):143-9.

22. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, et al. Human

skin Langerhans cells are targets of dengue virus infection. Nat Med. 2000;6(7):816-20.

23. Martina BE, Koraka P, Osterhaus AD. Dengue virus pathogenesis: an integrated view.

Clin Microbiol Rev. 2009;22(4):564-81. PMCID: 2772360.

24. Blackley S, Kou Z, Chen H, Quinn M, Rose RC, Schlesinger JJ, et al. Primary human

splenic macrophages, but not T or B cells, are the principal target cells for dengue virus infection

in vitro. J Virol. 2007;81(24):13325-34. PMCID: 2168870.

25. Boonnak K, Slike BM, Burgess TH, Mason RM, Wu SJ, Sun P, et al. Role of dendritic

cells in antibody-dependent enhancement of dengue virus infection. J Virol. 2008;82(8):3939-51.

PMCID: 2292981.

26. Ho LJ, Wang JJ, Shaio MF, Kao CL, Chang DM, Han SW, et al. Infection of human

dendritic cells by dengue virus causes cell maturation and cytokine production. J Immunol.

2001;166(3):1499-506.

27. Kwan WH, Helt AM, Maranon C, Barbaroux JB, Hosmalin A, Harris E, et al. Dendritic

cell precursors are permissive to dengue virus and human immunodeficiency virus infection. J

Virol. 2005;79(12):7291-9. PMCID: 1143643.

28. Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization of dengue virus in

naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis.

2004;189(8):1411-8.

Page 115: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

101

29. Balsitis SJ, Coloma J, Castro G, Alava A, Flores D, McKerrow JH, et al. Tropism of

dengue virus in mice and humans defined by viral nonstructural protein 3-specific

immunostaining. Am J Trop Med Hyg. 2009;80(3):416-24.

30. Huerre MR, Lan NT, Marianneau P, Hue NB, Khun H, Hung NT, et al. Liver

histopathology and biological correlates in five cases of fatal dengue fever in Vietnamese

children. Virchows Arch. 2001;438(2):107-15.

31. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, et al.

Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease

severity. J Infect Dis. 2000;181(1):2-9.

32. Murgue B, Roche C, Chungue E, Deparis X. Prospective study of the duration and

magnitude of viraemia in children hospitalised during the 1996-1997 dengue-2 outbreak in

French Polynesia. J Med Virol. 2000;60(4):432-8.

33. de Macedo FC, Nicol AF, Cooper LD, Yearsley M, Pires AR, Nuovo GJ. Histologic,

viral, and molecular correlates of dengue fever infection of the liver using highly sensitive

immunohistochemistry. Diagn Mol Pathol. 2006;15(4):223-8.

34. Halstead SB, Simasthien P. Observations related to the pathogenesis of dengue

hemorrhagic fever. II. Antigenic and biologic properties of dengue viruses and their association

with disease response in the host. Yale J Biol Med. 1970;42(5):276-92. PMCID: 2591694.

35. Halstead SB, Nimmannitya S, Cohen SN. Observations related to pathogenesis of dengue

hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered.

Yale J Biol Med. 1970;42(5):311-28. PMCID: 2591704.

36. Henchal EA, Putnak JR. The dengue viruses. Clin Microbiol Rev. 1990;3(4):376-96.

PMCID: 358169.

37. Rico-Hesse R. Molecular evolution and distribution of dengue viruses type 1 and 2 in

nature. Virology. 1990;174(2):479-93.

38. Gaunt MW, Sall AA, de Lamballerie X, Falconar AK, Dzhivanian TI, Gould EA.

Phylogenetic relationships of flaviviruses correlate with their epidemiology, disease association

and biogeography. J Gen Virol. 2001;82(Pt 8):1867-76.

39. Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, Lenches E, et al. Structure of

dengue virus: implications for flavivirus organization, maturation, and fusion. Cell.

2002;108(5):717-25.

40. Kaufmann B, Rossmann MG. Molecular mechanisms involved in the early steps of

flavivirus cell entry. Microbes Infect. 2011;13(1):1-9. PMCID: 3014442.

41. Zhang Y, Corver J, Chipman PR, Zhang W, Pletnev SV, Sedlak D, et al. Structures of

immature flavivirus particles. Embo J. 2003;22(11):2604-13. PMCID: 156766.

Page 116: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

102

42. Wengler G, Gross HJ. Studies on virus-specific nucleic acids synthesized in vertebrate

and mosquito cells infected with flaviviruses. Virology. 1978;89(2):423-37.

43. Markoff L. 5'- and 3'-noncoding regions in flavivirus RNA. Adv Virus Res. 2003;59:177-

228.

44. Falgout B, Chanock R, Lai CJ. Proper processing of dengue virus nonstructural

glycoprotein NS1 requires the N-terminal hydrophobic signal sequence and the downstream

nonstructural protein NS2a. J Virol. 1989;63(5):1852-60. PMCID: 250595.

45. Falgout B, Markoff L. Evidence that flavivirus NS1-NS2A cleavage is mediated by a

membrane-bound host protease in the endoplasmic reticulum. J Virol. 1995;69(11):7232-43.

PMCID: 189645.

46. Jones CT, Ma L, Burgner JW, Groesch TD, Post CB, Kuhn RJ. Flavivirus capsid is a

dimeric alpha-helical protein. J Virol. 2003;77(12):7143-9. PMCID: 156156.

47. Ma L, Jones CT, Groesch TD, Kuhn RJ, Post CB. Solution structure of dengue virus

capsid protein reveals another fold. Proc Natl Acad Sci U S A. 2004;101(10):3414-9. PMCID:

373476.

48. Stiasny K, Heinz FX. Flavivirus membrane fusion. J Gen Virol. 2006;87(Pt 10):2755-66.

49. Modis Y, Ogata S, Clements D, Harrison SC. Structure of the dengue virus envelope

protein after membrane fusion. Nature. 2004;427(6972):313-9.

50. Modis Y, Ogata S, Clements D, Harrison SC. Variable surface epitopes in the crystal

structure of dengue virus type 3 envelope glycoprotein. J Virol. 2005;79(2):1223-31. PMCID:

538574.

51. Zhang W, Chipman PR, Corver J, Johnson PR, Zhang Y, Mukhopadhyay S, et al.

Visualization of membrane protein domains by cryo-electron microscopy of dengue virus. Nat

Struct Biol. 2003;10(11):907-12.

52. Crabtree MB, Kinney RM, Miller BR. Deglycosylation of the NS1 protein of dengue 2

virus, strain 16681: construction and characterization of mutant viruses. Arch Virol.

2005;150(4):771-86.

53. Munoz-Jordan JL, Sanchez-Burgos GG, Laurent-Rolle M, Garcia-Sastre A. Inhibition of

interferon signaling by dengue virus. Proc Natl Acad Sci U S A. 2003;100(24):14333-8. PMCID:

283592.

54. Luo D, Xu T, Hunke C, Gruber G, Vasudevan SG, Lescar J. Crystal structure of the NS3

protease-helicase from dengue virus. J Virol. 2008;82(1):173-83. PMCID: 2224403.

55. Yap TL, Xu T, Chen YL, Malet H, Egloff MP, Canard B, et al. Crystal structure of the

dengue virus RNA-dependent RNA polymerase catalytic domain at 1.85-angstrom resolution. J

Virol. 2007;81(9):4753-65. PMCID: 1900186.

Page 117: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

103

56. Egloff MP, Benarroch D, Selisko B, Romette JL, Canard B. An RNA cap (nucleoside-2'-

O-)-methyltransferase in the flavivirus RNA polymerase NS5: crystal structure and functional

characterization. Embo J. 2002;21(11):2757-68. PMCID: 125380.

57. Zhou Y, Ray D, Zhao Y, Dong H, Ren S, Li Z, et al. Structure and function of flavivirus

NS5 methyltransferase. J Virol. 2007;81(8):3891-903. PMCID: 1866096.

58. Marovich M, Grouard-Vogel G, Louder M, Eller M, Sun W, Wu SJ, et al. Human

dendritic cells as targets of dengue virus infection. J Investig Dermatol Symp Proc.

2001;6(3):219-24.

59. Tassaneetrithep B, Burgess TH, Granelli-Piperno A, Trumpfheller C, Finke J, Sun W, et

al. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med.

2003;197(7):823-9. PMCID: 2193896.

60. Mukhopadhyay S, Kuhn RJ, Rossmann MG. A structural perspective of the flavivirus life

cycle. Nat Rev Microbiol. 2005;3(1):13-22.

61. Chu JJ, Rajamanonmani R, Li J, Bhuvanakantham R, Lescar J, Ng ML. Inhibition of

West Nile virus entry by using a recombinant domain III from the envelope glycoprotein. J Gen

Virol. 2005;86(Pt 2):405-12.

62. Huerta V, Chinea G, Fleitas N, Sarria M, Sanchez J, Toledo P, et al. Characterization of

the interaction of domain III of the envelope protein of dengue virus with putative receptors from

CHO cells. Virus Res. 2008;137(2):225-34.

63. Thepparit C, Smith DR. Serotype-specific entry of dengue virus into liver cells:

identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus

serotype 1 receptor. J Virol. 2004;78(22):12647-56. PMCID: 525075.

64. Cabrera-Hernandez A, Thepparit C, Suksanpaisan L, Smith DR. Dengue virus entry into

liver (HepG2) cells is independent of hsp90 and hsp70. J Med Virol. 2007;79(4):386-92.

65. Jindadamrongwech S, Thepparit C, Smith DR. Identification of GRP 78 (BiP) as a liver

cell expressed receptor element for dengue virus serotype 2. Arch Virol. 2004;149(5):915-27.

66. Das S, Laxminarayana SV, Chandra N, Ravi V, Desai A. Heat shock protein 70 on

Neuro2a cells is a putative receptor for Japanese encephalitis virus. Virology. 2009;385(1):47-57.

67. Salas-Benito J, Reyes-Del Valle J, Salas-Benito M, Ceballos-Olvera I, Mosso C, del

Angel RM. Evidence that the 45-kD glycoprotein, part of a putative dengue virus receptor

complex in the mosquito cell line C6/36, is a heat-shock related protein. Am J Trop Med Hyg.

2007;77(2):283-90.

68. van der Schaar HM, Rust MJ, Chen C, van der Ende-Metselaar H, Wilschut J, Zhuang X,

et al. Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells.

PLoS Pathog. 2008;4(12):e1000244. PMCID: 2592694.

Page 118: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

104

69. Acosta EG, Castilla V, Damonte EB. Functional entry of dengue virus into Aedes

albopictus mosquito cells is dependent on clathrin-mediated endocytosis. J Gen Virol. 2008;89(Pt

2):474-84.

70. Allison SL, Schalich J, Stiasny K, Mandl CW, Heinz FX. Mutational evidence for an

internal fusion peptide in flavivirus envelope protein E. J Virol. 2001;75(9):4268-75. PMCID:

114172.

71. Medigeshi GR, Hirsch AJ, Streblow DN, Nikolich-Zugich J, Nelson JA. West Nile virus

entry requires cholesterol-rich membrane microdomains and is independent of alphavbeta3

integrin. J Virol. 2008;82(11):5212-9. PMCID: 2395215.

72. Stiasny K, Fritz R, Pangerl K, Heinz FX. Molecular mechanisms of flavivirus membrane

fusion. Amino Acids. 2011;41(5):1159-63.

73. Harrison SC. The pH sensor for flavivirus membrane fusion. J Cell Biol.

2008;183(2):177-9. PMCID: 2568014.

74. Fritz R, Stiasny K, Heinz FX. Identification of specific histidines as pH sensors in

flavivirus membrane fusion. J Cell Biol. 2008;183(2):353-61. PMCID: 2568029.

75. Stiasny K, Kossl C, Lepault J, Rey FA, Heinz FX. Characterization of a structural

intermediate of flavivirus membrane fusion. PLoS Pathog. 2007;3(2):e20. PMCID: 1797619.

76. Kaufmann B, Chipman PR, Holdaway HA, Johnson S, Fremont DH, Kuhn RJ, et al.

Capturing a flavivirus pre-fusion intermediate. PLoS Pathog. 2009;5(11):e1000672. PMCID:

2776511.

77. Cleaves GR, Ryan TE, Schlesinger RW. Identification and characterization of type 2

dengue virus replicative intermediate and replicative form RNAs. Virology. 1981;111(1):73-83.

78. Bartenschlager R, Miller S. Molecular aspects of Dengue virus replication. Future

Microbiol. 2008;3(2):155-65.

79. Yu IM, Zhang W, Holdaway HA, Li L, Kostyuchenko VA, Chipman PR, et al. Structure

of the immature dengue virus at low pH primes proteolytic maturation. Science.

2008;319(5871):1834-7.

80. Li L, Lok SM, Yu IM, Zhang Y, Kuhn RJ, Chen J, et al. The flavivirus precursor

membrane-envelope protein complex: structure and maturation. Science. 2008;319(5871):1830-4.

81. Zhang Y, Zhang W, Ogata S, Clements D, Strauss JH, Baker TS, et al. Conformational

changes of the flavivirus E glycoprotein. Structure. 2004;12(9):1607-18.

82. Abbas A. Cellular and Molecular Immunology 2012.

83. Murphy KM. Janeway's Immunobiology2012.

Page 119: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

105

84. Katze MG, He Y, Gale M, Jr. Viruses and interferon: a fight for supremacy. Nat Rev

Immunol. 2002;2(9):675-87.

85. Weber F, Kochs G, Haller O. Inverse interference: how viruses fight the interferon

system. Viral Immunol. 2004;17(4):498-515.

86. Morrison J, Aguirre S, Fernandez-Sesma A. Innate immunity evasion by Dengue virus.

Viruses. 2012;4(3):397-413. PMCID: 3347034.

87. Samuel CE. Antiviral actions of interferons. Clin Microbiol Rev. 2001;14(4):778-809,

table of contents. PMCID: 89003.

88. Mitchell DA, Fadden AJ, Drickamer K. A novel mechanism of carbohydrate recognition

by the C-type lectins DC-SIGN and DC-SIGNR. Subunit organization and binding to multivalent

ligands. J Biol Chem. 2001;276(31):28939-45.

89. Heil F, Ahmad-Nejad P, Hemmi H, Hochrein H, Ampenberger F, Gellert T, et al. The

Toll-like receptor 7 (TLR7)-specific stimulus loxoribine uncovers a strong relationship within the

TLR7, 8 and 9 subfamily. Eur J Immunol. 2003;33(11):2987-97.

90. Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, et al. Recognition of

single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A.

2004;101(15):5598-603. PMCID: 397437.

91. Navarro-Sanchez E, Despres P, Cedillo-Barron L. Innate immune responses to dengue

virus. Arch Med Res. 2005;36(5):425-35.

92. Della Chiesa M, Sivori S, Castriconi R, Marcenaro E, Moretta A. Pathogen-induced

private conversations between natural killer and dendritic cells. Trends Microbiol.

2005;13(3):128-36.

93. Bashyam HS, Green S, Rothman AL. Dengue virus-reactive CD8+ T cells display

quantitative and qualitative differences in their response to variant epitopes of heterologous viral

serotypes. J Immunol. 2006;176(5):2817-24.

94. Duangchinda T, Dejnirattisai W, Vasanawathana S, Limpitikul W, Tangthawornchaikul

N, Malasit P, et al. Immunodominant T-cell responses to dengue virus NS3 are associated with

DHF. Proc Natl Acad Sci U S A. 2010;107(39):16922-7. PMCID: 2947904.

95. Gunther VJ, Putnak R, Eckels KH, Mammen MP, Scherer JM, Lyons A, et al. A human

challenge model for dengue infection reveals a possible protective role for sustained interferon

gamma levels during the acute phase of illness. Vaccine. 2011;29(22):3895-904.

96. Mangada MM, Rothman AL. Altered cytokine responses of dengue-specific CD4+ T

cells to heterologous serotypes. J Immunol. 2005;175(4):2676-83.

97. Kurane I, Meager A, Ennis FA. Dengue virus-specific human T cell clones. Serotype

crossreactive proliferation, interferon gamma production, and cytotoxic activity. J Exp Med.

1989;170(3):763-75. PMCID: 2189437.

Page 120: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

106

98. Vaughan K, Greenbaum J, Blythe M, Peters B, Sette A. Meta-analysis of all immune

epitope data in the Flavivirus genus: inventory of current immune epitope data status in the

context of virus immunity and immunopathology. Viral Immunol. 2010;23(3):259-84. PMCID:

2942863.

99. Yauch LE, Zellweger RM, Kotturi MF, Qutubuddin A, Sidney J, Peters B, et al. A

protective role for dengue virus-specific CD8+ T cells. J Immunol. 2009;182(8):4865-73.

PMCID: 2674070.

100. Valdes K, Alvarez M, Pupo M, Vazquez S, Rodriguez R, Guzman MG. Human Dengue

antibodies against structural and nonstructural proteins. Clin Diagn Lab Immunol. 2000;7(5):856-

7. PMCID: 95972.

101. Churdboonchart V, Bhamarapravati N, Peampramprecha S, Sirinavin S. Antibodies

against dengue viral proteins in primary and secondary dengue hemorrhagic fever. Am J Trop

Med Hyg. 1991;44(5):481-93.

102. Hatch S, Endy TP, Thomas S, Mathew A, Potts J, Pazoles P, et al. Intracellular cytokine

production by dengue virus-specific T cells correlates with subclinical secondary infection. J

Infect Dis. 2011;203(9):1282-91. PMCID: 3069729.

103. Schieffelin JS, Costin JM, Nicholson CO, Orgeron NM, Fontaine KA, Isern S, et al.

Neutralizing and non-neutralizing monoclonal antibodies against dengue virus E protein derived

from a naturally infected patient. Virol J. 2010;7:28. PMCID: 2829534.

104. Roehrig JT, Bolin RA, Kelly RG. Monoclonal antibody mapping of the envelope

glycoprotein of the dengue 2 virus, Jamaica. Virology. 1998;246(2):317-28.

105. Wahala WM, Kraus AA, Haymore LB, Accavitti-Loper MA, de Silva AM. Dengue virus

neutralization by human immune sera: role of envelope protein domain III-reactive antibody.

Virology. 2009;392(1):103-13. PMCID: 2746956.

106. Sukupolvi-Petty S, Austin SK, Engle M, Brien JD, Dowd KA, Williams KL, et al.

Structure and function analysis of therapeutic monoclonal antibodies against dengue virus type 2.

J Virol. 2010;84(18):9227-39. PMCID: 2937608.

107. Beltramello M, Williams KL, Simmons CP, Macagno A, Simonelli L, Quyen NT, et al.

The human immune response to Dengue virus is dominated by highly cross-reactive antibodies

endowed with neutralizing and enhancing activity. Cell Host Microbe. 2010;8(3):271-83.

108. Mehlhop E, Ansarah-Sobrinho C, Johnson S, Engle M, Fremont DH, Pierson TC, et al.

Complement protein C1q inhibits antibody-dependent enhancement of flavivirus infection in an

IgG subclass-specific manner. Cell Host Microbe. 2007;2(6):417-26. PMCID: 2194657.

109. Shaio MF, Chang FY, Hou SC. Complement pathway activity in serum from patients

with classical dengue fever. Trans R Soc Trop Med Hyg. 1992;86(6):672-5.

Page 121: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

107

110. Lei HY, Yeh TM, Liu HS, Lin YS, Chen SH, Liu CC. Immunopathogenesis of dengue

virus infection. J Biomed Sci. 2001;8(5):377-88.

111. Green S, Vaughn DW, Kalayanarooj S, Nimmannitya S, Suntayakorn S, Nisalak A, et al.

Early immune activation in acute dengue illness is related to development of plasma leakage and

disease severity. J Infect Dis. 1999;179(4):755-62.

112. Endy TP, Nisalak A, Chunsuttitwat S, Vaughn DW, Green S, Ennis FA, et al.

Relationship of preexisting dengue virus (DV) neutralizing antibody levels to viremia and

severity of disease in a prospective cohort study of DV infection in Thailand. J Infect Dis.

2004;189(6):990-1000.

113. An J, Zhou DS, Zhang JL, Morida H, Wang JL, Yasui K. Dengue-specific CD8+ T cells

have both protective and pathogenic roles in dengue virus infection. Immunol Lett.

2004;95(2):167-74.

114. Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke DS. Antibody-dependent enhancement

of dengue virus growth in human monocytes as a risk factor for dengue hemorrhagic fever. Am J

Trop Med Hyg. 1989;40(4):444-51.

115. Halstead SB. Observations related to pathogensis of dengue hemorrhagic fever. VI.

Hypotheses and discussion. Yale J Biol Med. 1970;42(5):350-62. PMCID: 2591710.

116. Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that maternal dengue

antibodies are important in the development of dengue hemorrhagic fever in infants. Am J Trop

Med Hyg. 1988;38(2):411-9.

117. Selin LK, Cornberg M, Brehm MA, Kim SK, Calcagno C, Ghersi D, et al. CD8 memory

T cells: cross-reactivity and heterologous immunity. Semin Immunol. 2004;16(5):335-47.

118. Blaney JE, Jr., Durbin AP, Murphy BR, Whitehead SS. Development of a live attenuated

dengue virus vaccine using reverse genetics. Viral Immunol. 2006;19(1):10-32.

119. Sun W, Nisalak A, Gettayacamin M, Eckels KH, Putnak JR, Vaughn DW, et al.

Protection of Rhesus monkeys against dengue virus challenge after tetravalent live attenuated

dengue virus vaccination. J Infect Dis. 2006;193(12):1658-65.

120. Huang CY, Butrapet S, Pierro DJ, Chang GJ, Hunt AR, Bhamarapravati N, et al.

Chimeric dengue type 2 (vaccine strain PDK-53)/dengue type 1 virus as a potential candidate

dengue type 1 virus vaccine. J Virol. 2000;74(7):3020-8. PMCID: 111800.

121. Butrapet S, Huang CY, Pierro DJ, Bhamarapravati N, Gubler DJ, Kinney RM.

Attenuation markers of a candidate dengue type 2 vaccine virus, strain 16681 (PDK-53), are

defined by mutations in the 5' noncoding region and nonstructural proteins 1 and 3. J Virol.

2000;74(7):3011-9. PMCID: 111799.

122. Sanchez V, Gimenez S, Tomlinson B, Chan PK, Thomas GN, Forrat R, et al. Innate and

adaptive cellular immunity in flavivirus-naive human recipients of a live-attenuated dengue

serotype 3 vaccine produced in Vero cells (VDV3). Vaccine. 2006;24(23):4914-26.

Page 122: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

108

123. McArthur JH, Durbin AP, Marron JA, Wanionek KA, Thumar B, Pierro DJ, et al. Phase I

clinical evaluation of rDEN4Delta30-200,201: a live attenuated dengue 4 vaccine candidate

designed for decreased hepatotoxicity. Am J Trop Med Hyg. 2008;79(5):678-84. PMCID:

2590927.

124. Guirakhoo F, Pugachev K, Arroyo J, Miller C, Zhang ZX, Weltzin R, et al. Viremia and

immunogenicity in nonhuman primates of a tetravalent yellow fever-dengue chimeric vaccine:

genetic reconstructions, dose adjustment, and antibody responses against wild-type dengue virus

isolates. Virology. 2002;298(1):146-59.

125. Huang CY, Butrapet S, Tsuchiya KR, Bhamarapravati N, Gubler DJ, Kinney RM.

Dengue 2 PDK-53 virus as a chimeric carrier for tetravalent dengue vaccine development. J

Virol. 2003;77(21):11436-47. PMCID: 229366.

126. Kochel T, Wu SJ, Raviprakash K, Hobart P, Hoffman S, Porter K, et al. Inoculation of

plasmids expressing the dengue-2 envelope gene elicit neutralizing antibodies in mice. Vaccine.

1997;15(5):547-52.

127. Men R, Wyatt L, Tokimatsu I, Arakaki S, Shameem G, Elkins R, et al. Immunization of

rhesus monkeys with a recombinant of modified vaccinia virus Ankara expressing a truncated

envelope glycoprotein of dengue type 2 virus induced resistance to dengue type 2 virus challenge.

Vaccine. 2000;18(27):3113-22.

128. Raviprakash K, Wang D, Ewing D, Holman DH, Block K, Woraratanadharm J, et al. A

tetravalent dengue vaccine based on a complex adenovirus vector provides significant protection

in rhesus monkeys against all four serotypes of dengue virus. J Virol. 2008;82(14):6927-34.

PMCID: 2446963.

129. Naniche D, Varior-Krishnan G, Cervoni F, Wild TF, Rossi B, Rabourdin-Combe C, et al.

Human membrane cofactor protein (CD46) acts as a cellular receptor for measles virus. J Virol.

1993;67(10):6025-32. PMCID: 238023.

130. Erlenhoefer C, Wurzer WJ, Loffler S, Schneider-Schaulies S, ter Meulen V, Schneider-

Schaulies J. CD150 (SLAM) is a receptor for measles virus but is not involved in viral contact-

mediated proliferation inhibition. J Virol. 2001;75(10):4499-505. PMCID: 114203.

131. Muhlebach MD, Mateo M, Sinn PL, Prufer S, Uhlig KM, Leonard VH, et al. Adherens

junction protein nectin-4 is the epithelial receptor for measles virus. Nature. 2011;480(7378):530-

3. PMCID: 3245798.

132. del Valle JR, Devaux P, Hodge G, Wegner NJ, McChesney MB, Cattaneo R. A vectored

measles virus induces hepatitis B surface antigen antibodies while protecting macaques against

measles virus challenge. J Virol. 2007;81(19):10597-605. PMCID: 2045491.

133. Despres P, Combredet C, Frenkiel MP, Lorin C, Brahic M, Tangy F. Live measles

vaccine expressing the secreted form of the West Nile virus envelope glycoprotein protects

against West Nile virus encephalitis. J Infect Dis. 2005;191(2):207-14.

Page 123: Expression of Dengue virus envelope glycoproteins …...Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector by Rowida Abdelgalel A Thesis Presented in

109

134. Brandler S, Ruffie C, Najburg V, Frenkiel MP, Bedouelle H, Despres P, et al. Pediatric

measles vaccine expressing a dengue tetravalent antigen elicits neutralizing antibodies against all

four dengue viruses. Vaccine. 2010;28(41):6730-9.

135. Reyes-del Valle J, de la Fuente C, Turner MA, Springfeld C, Apte-Sengupta S, Frenzke

ME, et al. Broadly neutralizing immune responses against hepatitis C virus induced by vectored

measles viruses and a recombinant envelope protein booster. J Virol. 2012;86(21):11558-66.

PMCID: 3486281.

136. WHO. Dengue Fever and Dengue Haemorrhagic Fever. 2013 [updated 2013; cited 2013

11/25/2013]; Available from:

http://www.searo.who.int/entity/vector_borne_tropical_diseases/en/index.html.

137. Weiss BG, Schlesinger S. Recombination between Sindbis virus RNAs. J Virol.

1991;65(8):4017-25. PMCID: 248832.

138. Seligman SJ, Gould EA. Live flavivirus vaccines: reasons for caution. Lancet.

2004;363(9426):2073-5.

139. Brandler S, Tangy F. Recombinant vector derived from live attenuated measles virus:

potential for flavivirus vaccines. Comp Immunol Microbiol Infect Dis. 2008;31(2-3):271-91.

140. Brandler S, Lucas-Hourani M, Moris A, Frenkiel MP, Combredet C, Fevrier M, et al.

Pediatric measles vaccine expressing a dengue antigen induces durable serotype-specific

neutralizing antibodies to dengue virus. PLoS Negl Trop Dis. 2007;1(3):e96. PMCID: 2154386.

141. Watanabe S, Shirogane Y, Suzuki SO, Ikegame S, Koga R, Yanagi Y. Mutant fusion

proteins with enhanced fusion activity promote measles virus spread in human neuronal cells and

brains of suckling hamsters. J Virol. 2013;87(5):2648-59. PMCID: 3571373.