siRNA BASED INHIBITION OF DENGUE VIRUS AND

PREDICTION OF POSSIBLE VACCINAL TARGETS

UmmarRaheel

2010-NUST-TfrPhD-V&I-44

Atta-ur-Rahman School of Applied Biosciences National University of Sciences & Technology Islamabad Pakistan 2015 siRNA BASED INHIBITION OF DENGUE VIRUS AND

PREDICTION OF POSSIBLE VACCINAL TARGETS

UmmarRaheel

A thesis submitted in partial fulfillment of the requirement for the

degree of Doctor of Philosophy in

Virology and Immunology

Supervision

Supervisor

Dr.Najam Us SaharSadafZaidi

Atta-ur-Rahman School of Applied Biosciences National University of Sciences & Technology IslamabadPakistan 2015

Dedicated to

My Almighty Allah and my Mother…

For providing me strength and patience

Acknowledgements

ACKNOWLEDGMENTS

All praises for almighty Allah the creator of the universe. Who created human mind and give us the power to conceive ideas and make decisions. Who guided us to follow the right pathway and without his guidance we were blind and ignorant.

I am extremely thankful to my supervisor Dr.SadafZaidi for her never ending help, guidance and patience throughout my studies. I am highly thankful to National

University of Sciences and technology and Higher Education Commission for providing me funding for my research. I am thankful to Dr. Muhammad Tahir, Dr.HajraSadia and

Dr.RoquyyaGul (UOL) my thesis committee members, for their support and guidance. I am also thankful to Dr. Peter John and Dr.AttyaBhatti for their support during my study.

I owe immense acknowledgement to Prof AravindaDesilva, Department of

Microbiology and Immunology, University of Chapel Hill North Carolina, USA for accepting me in his lab as visiting research scholar. I did most of the research work in his lab. He allowed me to utilize all available facilities for my research work in addition with constant guidance and support.

During my PhD research work, I met many amazing people who were my research colleagues and great friends and I knew I could rely on them whenever I needed.

My research work would not have been possible without the assistance of Dr Muhammad

Imran, Dr Mushin Jamal, NasirRaiz, Muhammad Fahim, IrshadulHaq, HashaamAkhatar and every member of AttaUr Rahman School of Applied Biosciences.

v

Acknowledgements

I can never forget the role of my mother who inspired me and prayed for my success when I was down and she did not let me fall. I thank Allah every day for blessing me with a great mother. I must acknowledge my Fiancée for her support during the last two years; it was her constant encouragement that made me withstand difficulties and disappointments. I will never forget the role of my family in reaching this milestone.

Finally I would again thank Dr.SadafZaidi for being a great person who was not only my supervisor in research but also a source of inspiration.

UmmarRaheel

vi

Table of Contents

TABLE OF CONTENTS

Title Page #

Acknowledgements v

List of Abbreviations vii

List of Tables xii

List of Figures xiii

Abstract xvii

Chapter -1

Introduction 01

1.1 Dengue virus 01

1.1.1 Epidemiology 02

1.2 DENV infection in Pakistan 04

1.3 DENV genome 05

1.4 Replication and life cycle of DENV 06

1.5 Pathogenesis 08

1.6 In vitro culturing 09

1.7 Animal models 10

1.8 RNA interference 11

1.9 Insilico analysis 12

1.10 Aim and objectives 13

vii

Table of Contents

Chapter -2

Review of Literature 23

2.1 Genome organization of 24

2.2 DENV proteins 25

2.3 Dengue virus 31

2.4 DENV variations 33

2.5 Pathogenesis 34

2.5.1 High virulence 34

2.5.2 Host innate immunity 35

2.5.3 Antibody dependent autoimmune responses in DF 35

2.5.4 Cellular autoimmunity in DHF/DSS 36

2.5.5 Antibody Associated with Enhancement of DF 36

2.6 Viral targets for DENV inhibitors 38

2.6.1 Structural proteins’ based inhibition of DENV 38

2.6.2 Nucleoside based inhibition of DENV 39

2.6.3 Methyltransferases inhibition of DENV 39

2.6.4 Protease (NS2B/NS3) inhibition of DENV 41

2.6.5 Host enzymes based inhibition of DENV 42

2.6.6 Kinases based inhibition of DENV 43

2.6.7 Glucosidase based inhibition of DENV 43

2.6.8 Cytokines and receptors based inhibition of DENV 44

2.6.9 Computational design of peptide inhibitors against DENV 45

viii

Table of Contents

2.7 Inhibition of DENV by siRNA 45

2.8 Vaccine development for DENV 47

2.8.1Chimeric vaccine 48

2.8.2 DNA vaccine 48

2.8.3 Live attenuated vaccines 49

2.8.4 Subunit vaccine 50

2.9 Epitope variation a challenge for DENV vaccine design 51

2.9.1DENV T Cell antigenic diversity analysis 52

2.9.2 T cell epitope mapping of DENV 52

2.9.3 Insilico analysis of DENV T cell epitopes 53

2.9.4 B cell epitope mapping antigenic diversity analysis 53

2.9.45 Insilico analysis of DENV B cell epitopes 54

Chapter -3

Materials and Methods 57

3.1 SerotypessiRNA designing/ bioinformatics 57

3.2 Mammalian cells and DENV 57

3.3 siRNA transfections of cell cultures 58

3.4 Focus assay 58

3.5 Immunofluorescence Assay (IFA) 59

3.6 Western blot analysis 60

3.7 Realtime PCR analysis of DENV RNA 60

ix

Table of Contents

3.8.1 RNA extraction for qPCR 60

3.8.2 Optimizing of qPCR 61

3.9 Animal model of DENV2 61

3.9.1 Intracerebral injection 61

3.9.2 RNA Extraction from different tissues 62

3.9.3 Primer Designing for DENV RNA detection 63

3.9.4 Complementary DNA (cDNA) synthesis 64

3.9.5 Polymerase chain reaction 64

3.9.6 Purification of PCR products 64

3.9.7 Ligation 65

3.9.9 Transformation 65

3.10 Screening and selection of clones 66

3.10.1 Colony PCR 66

3.10.2 Plasmid DNA isolation and restriction digestion 67

3.11 Sequencing 68

3.12 Prediction of vaccine targets 68

3.12.1 DENV2 sequence comparison and phylogenetic analysis 68

3.12.2 Predication of epitopes on DENV2 Pakistani Isolate 68

3.13 Insilicomodeling of DENV2 structural proteins from Pakistani isolate 69

Chapter-4

x

Table of Contents

Results 70

4.1 Nucleotide analysis of un-translated terminal repeats 70

4.1.1 Alignment results for 3'UTR of DENV2 70

4.1.2 Alignment results for 3'UTR of DENV3 73

4.1.3 Alignment results for 5'UTR of DENV2 77

4.1.4Alignment results for 5'UTR of DENV3 80

4.1.5Alignment results for structural region of DENV2 genome 90

4.1.6 Alignment results for structural region of DENV3 genome 99

4.2 siRNAdesigning and bioinformatics 89

4.3 Focus assay 102

4.4Immunofloroscence assay 107

4.5 Western blotting 120

4.9 Cloning, sequencing of DENV2 structural genes 131

4.9 DENV2 structural genes phylogenetic analysis 141

4.10 Full length epitopic sites prediction of Pakistani DENV2 structural proteins 145

4.11 Full length B cell epitopic sites prediction of Pakistani DENV2 capsid protein 146

4.12 Full length epitopic sites prediction of Pakistani DENV2 prM protein 153

4.13 Full Length epitopic Sites Prediction of Pakistani DENV2 E protein 160

4.14 DENV2 structural proteins homology modeling 167

Chapter -5

xi

Table of Contents

Discussion 174

Chapter -6

References 185

xii

List of Tables

LIST OF TABLES

Table 2.1DENV RNA elements and there functions 30

Table 2.2 DENV proteins and there functions 31

Table 3.1 Primers for detection of DENV2 structural genes 63

Table 4.1 Designed siRNAs and their location in DENV2 genome 100

Table 4.2 Designed siRNAs and their location in DENV3 genome 101

Table 4.3 Focus assay triplet experiment for DENV2 load estimation 103

Table 4.4 Focus assay triplet experiment for DENV3 load estimation. 105

xii

List of Figures

LIST OF FIGURES

Figure # Title Page #

1.1 Epidemiology of DENV 03

1.2 Replication cycle of DENV 06

1.3 Genome organization of DENV 09

1.4 DENV RNA elements and proteins 11

1.5 RNA interference pathways 30

4.1 Sequence analysis of DENV2 3'UTR 72

4.2 Sequence analysis of DENV3 3'UTR 76

4.3 Sequence analysis of DENV2 5'UTR 79

4.4 Sequence analysis of DENV3 5'UTR 81

4.5 Sequence analysis of DENV2 structural region 89

4.6 Sequence analysis of DENV3 structural region 98

4.7 Focus assay of siRNADENV2 transfected Vero-81 cells 104

4.8 Focus assay of siRNADENV3 transfected Vero-81 cells 106

4.9 IFA images of DENV2UTR5'siRNA1 treated Vero-81 cells 108

xiii

List of Figures

4.10 IFA images of DENV2UTR5'siRNA2 treated Vero-81 cells 109

4.11 IFA images of DENV2SsiRNA1 treated Vero-81 cells. 110

4.12 IFA images of DENV2SsiRNA2 treated Vero-81 cells. 111

4.13 IFA images of DENV2UTR3'siRNA1 treated Vero-81 cells 112

4.14 IFA images of DENV2UTR3'siRNA2 treated Vero-81 cells 113

4.15 IFA images of DENV3UTR5'siRNA1 treated Vero-81 cells 114

4.16 IFA images of DENV3UTR5'siRNA2 treated Vero-81 cells 115

4.17 IFA images of DENV3SsiRNA1 treated Vero-81 cells. 116

4.18 IFA images of DENV3SsiRNA2 treated Vero-81 cells. 117

4.19 IFA images of DENV3UTR3'siRNA1 treated Vero-81 cells 118

4.20 IFA images of DENV3UTR3'siRNA2 treated Vero-81 cells 119

4.21 Western blot analysis of mature and immature DENV2 121

4.22 Western blot analysis of siRNA transfected cells post- 122

DENV2 infection.

4.23 Western blot analysis of siRNA transfected cells post- 123

DENV3 infection.

4.24 Q-PCR for analysis of DENV2 RNA levels. 125

xiv

List of Figures

4.25 Q-PCR for checking DENV3 RNA levels. 126

4.26 Dissection of DENV2 mice model 127

4.27 Amplification of DENV2 C from different organs of mice. 129

4.28 Amplification of DENV2 prM from different organs of 130 mice.

4.29 Amplification of DENV2 C gene from local DENV2 isolate. 132

4.30 Colony PCR of DENV2 C gene. 133

4.31 DENV2 C clone confirmation by restriction digestion. 134

4.32 Amplification of DENV2 prM gene from local DENV2 135 isolate.

4.33 Colony PCR of DENV2 prM gene. 136

4.34 DENV2 prM clone confirmation by restriction digestion. 137

4.35 Amplification of DENV2 E gene from local DENV2 isolate 138

4.36 Colony PCR of DENV2 E gene. 139

4.37 DENV2 E clone confirmation by restriction digestion 140

4.38 Phylogenetic tree of DENV2 C gene 142

4.39 Phylogenetic tree of DENV2 prM gene. 143

4.40 Phylogenetic tree of DENV2 E gene. 144

xv

List of Figures

4.41 Bepipred linear epitope prediction of DENV2 C protein. 147

4.42 Chou and Fasman Beta-turn prediction of DENV2 C protein. 148

4.43 Emini surface accessibility prediction of DENV2 C protein. 149

4.44 Karplus and Schulz flexibility prediction of DENV2 C 150 protein.

4.45 Kolaskar and tongaonkar antigenicity of DENV2 C protein. 151

4.46 Parker hydrophilicity prediction of DENV2 C protein 152 DENV2 infection. 4.47 Bepipred linear epitope prediction DENV2 of prM. 154

4.48 Chou and Fasman Beta-turn prediction DENV2 of prM. 155

4.49 Emini surface accessibility prediction DENV2 of prM. 156

4.50 Karplus and Schulz flexibility prediction DENV2 of prM 157

4.51 Kolaskar and tongaonkar antigenicity prediction DENV2 of 158

prM.

4.52 Parker hydrophilicity prediction DENV2 of prM. 159

4.53 Bepipred linear epitope prediction of E protein. 161

4.54 Chou and Fasman Beta-turn prediction of E protein. 162

4.55 Emini surface accessibility prediction of E protein. 163

4.56 Karplus and Schulz flexibility prediction of E protein. 164

xvi

List of Figures

4.57 Kolaskar and tongaonkar antigenicity prediction of E 165

protein.

4.58 Parker hydrophilicity prediction of E protein 166

4.59 Phyre2 web server generated model of DENV2 C protein 168

represented by a colored 3D structure

4.60 Phyre2 web server generated model of DENV2 C protein 169

represented by a colored 3D structure

4.61 Phyre2 web server generated model of DENV2 prM protein 170

represented by a colored 3D structure

4.62 Phyre2 web server generated model of DENV2 prM protein 171

represented by a colored 3D structurewith epitopes

4.63 Phyre2 web server generated model of DENV2 E protein 172

represented by a colored 3D structure

4.64 Phyre2 web server generated model of DENV2 E protein 173

represented by a colored 3D structurewith epitopes

xvii

Abstract

ABSTRACT

Dengue virus (DENV) is an insect borne virus classified in the family

Flaviviridae. DENV is the most widespread arthropod borne virus across the globe and according to latest epidemiological reports dengue fever (DF) is one of the major cause of concern for global health.Amongst all four DENV serotypes predominately circulating serotypes in Pakistan are DENV2 and DENV3. RNA interference (siRNA and miRNA) is at the forefront of molecular approaches for inhibitory studies. The present study was aimed at inhibition of both these serotypes via siRNA interference; moreover structural genes were cloned for insilico analysisto facilitate strategies for the development of vaccines against DENV.

Both DENV2 and DENV3 were targeted by synthetic siRNAsdesigned against conserve regions in the genome. Six siRNAs were designed against both DENV2 and

DENV3 from three vital regions of genome 5'untranslated region (5'UTR), 3'UTR and structural genes. Transfections were performed by Lipofectamine and level of inhibition was observed by performing several viral titer estimation assays. Results showed thatDENV3UTR3'siRNA2 targeting 3' UTR was able to inhibit DENV3 replication.The replication cycle of DENV has revealed that in human Dendritic cells (DCs),Furin enzyme converts the immature DENV pre-membrane protein (prM) to mature M protein assisted by change in cellular pH.Therefore, in the later part of study fully mature

DENV2 were allowed to infect and later targeted by RNAi. Results from this study showed that siRNA (DENV2SsiRNA2) targeting the M region of genome was able to

xvi

Abstract

drastically knock down DENV2 in Vero-81 cells. Substantial inhibition was also seen by siRNA (DENV2UTR3'siRNA2) targeting 3' UTR region ofDENV2 genome.

Structural genes were cloned from locally isolated DENV2 serotype. All three genes (Capsid, prM and Envelope) were subjected to insilico analysis involving construction of phylogenetic trees and prediction of epitopic domains. It was observed that both Pakistani and Indian DENV2 share a strong sequence similarity. Inhibition of both DENV serotypes via RNAi highlights the effectiveness of molecular approaches for anti-dengue drug development. Moreover, findings of insilico study of Pakistani DENV2 serotype will assist in the development of regionally targeted vaccine for densely populated South Asian region.

xvii

Chapter 1 Introduction

Chapter 1

INTRODUCTION

1.1 DENGUE VIRUS

Dengue Virus (DENV) is a positive sense RNA virus which belongs to family

Flaviviridae, DENV has four serotypes (DENV1-4), is transmitted by mosquitoes

Aedesaegyptiand Aedesalbopictus(Rigau-Pérez et al., 1998). Amongst arthropods borne viruses DENV has highest rates in terms of annual cases and deaths, owing largely to growing population and travel(Lai et al., 1991). DENV has RNA genome which contains

5' cap on one side and 3' poly A tail (Gibbons and Vaughn, 2002). One of the reason for a lack of vaccine against DENV is antigenic difference amongst four serotype(Avirutnan et al., 2006).

DENV genome is positive sense single stranded RNA virus having a size of approximately 10.7 kb. The 5'end of genome known as untranslated terminal region

(5'UTR) is a100 nucleotide long stretch whereas genome ends at 3'UTR which has more than 400 nucleotides. Genome analysis revealed that there is a 5' cap at 5'UTR on the other hand 3'UTR is naked and lacks a polyadenylation at 3' end (Alvarez et al., 2005b).

UTRs are responsible for genome cycling, organization and replication for production of progeny DENVs. Several host cellular as well as viral factors interact with these UTRs for DENV replication (Alvarez et al., 2005a; Alvarez et al., 2008; Cui et al., 1998; De

Nova-Ocampo et al., 2002; Garcı́a-Montalvo et al., 2004). The region of genome towards the 5' end encodes structural proteins capsid (C), membrane (M) and envelope (E) at the

1

Chapter 1 Introduction

same time the rest of the portion of genome encodes nonstructural proteins (NS) (Rice et al., 1985).

1.1.1 EPIDEMIOLOGY

Worldwide DENV is prevalent in tropical and sub-tropical regions, urban populations of countries in Southeast Asia, South Asia and South America are at high risk(Lai et al., 1991). Dengue fever (DF) can be a self-limited or in some cases it can progress to sever conditions like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Both DHF and DSS are characterize by high fever, enhanced vascular permeability, thrombocytopenia, these complications can cause death in the absence of proper management (Avirutnan et al., 2006; Gibbons and Vaughn, 2002;Huang et al.,

2003).

According to WHO, DF was the most prevalent insect borne viral illness in 2012, due to the rising number of annual cases (Murray et al., 2013). Economic losses relating with DENV were mentioned in recent reports showing that cost associated with DF. In

South Asian and American countries was roughly US$3 billion (Shepard et al., 2013).

Although precise number of DF cases are still not known however, DENV has be isolated from 125 countries worldwide which is the indication of the magnitude of this global crisis (Ferreira, 2012).

In 1970s rigorous measures were taken for controlling mosquito populations for curbing Malaria and DF, even with these efforts sporadic break were recorded (Guzman et al., 1984a; Guzman et al., 1984b; Halstead, 1980). According to World Health

Organization (WHO) number of reported cases between 1950s to 1990s were approximately 2.5 million with more than 42000 deaths(Kautner et al., 1997). In the mid

2

Chapter 1 Introduction

1990s DF was confirmed in patients from 24 American countries, with many countries facing worst epidemics. In recent times first reported epidemic in Indian Sub-continent was reported in 1996. Since 2006 several DF epidemics were seen in South Asia, according to WHO's estimates in Pakistan most number of cases were reported in the outbreak of 2011 as shown in Figure 1.1.

Figure 1.1: Epidemiology of DENV: Geographical distribution of DF in the world and countries which are located in the risk zones.The global incidence of dengue fever in the year 2013 according to WHO estimates. The prevalence of dengue cases is depicted by brown color. (WHO report, 2013)

3

Chapter 1 Introduction

1.2 DENV INFECTION IN PAKISTAN

Dengue fever is endemic in Pakistani population and highest number of cases are reported during or post monsoon seasons (Jahan, 2011). During mid 1980s DF was reported for the first time in children which were having persistent high fever (Akram et al., 1998). The Baluchistan province was hit by a deadly DF outbreak in the year 1995 resulting in 57 deaths (Paul et al., 1998). With each passing year number of DF cases were rising and in 2003 a major outbreak resulted in approximately 1000 cases and 7 confirmed deaths in the Haripur region located in KPK province (Tang et al., 2008). One year later in 2004, more than 2500 confirmed DF cases were reported in the Punjab region and molecular analysis revealed that DENV2 was predominant serotype in both of these outbreaks.

One of the worst year for DF in Pakistan was 2006 in which 5400 cases were reported and 55 people lost their lives, however a large number of cases were not reported owing to the inadequate medical facilities. For this outbreak two serotypes which were held responsible were DENV2 and DENV3 respectively (Khan et al., 2007).

Lahore was severely affected by three circulating serotypes (DENV 2, 3 and 4) and resulted in 1800 DF cases in 2008. This co-circulation led to tremendous increase in the rate of DHF and resulted in several deaths. Country wide outbreak in the year 2010 resulted in more than 5000 confirm cases and several deaths, both DENV1 and DENV2 were present in the outbreak (Mahmood et al., 2012).

The largest recorded outbreak of DF occurred in 2011, with more than 20,000 countrywide confirmed cases and a staggering 300 deaths. The current trend show that

4

Chapter 1 Introduction

Pakistani population is facing a severe threat from DF and an outbreak can strike at any time. There is an urgent need for development of anti-dengue drug and vaccine in addition with measures for controlling vector population (Jahan, 2011).

1.3 DENV GENOME

The whole DENV poly-protein produced by the open reading frame (ORF) is in the following sequence 5'- C-prM/M-E-NS1-NS2A-NS2B-NS3-NS4A-NS4BNS5-3' as shown in Figure 1.3. The 3'end of DENV genome encodes non structural proteins, the proper roles of NS proteins is still not clear, however NS1 to N4A/B in association with

C protein are in the encapsidation of DENV RNA. Larger structural proteins M and E form a lipid bilayer structure which ultimately becomes envelope of new DENV. One of the important step in DENV maturation is cleavage of prMto M which occurs in the

Trans Golgi network. Furin is a pH dependent enzyme, when the pH of Golgi network drops this activates the cellular Furin resulting in cleavage of prM(Mackenzie and

Westaway, 2001).

5

Chapter 1 Introduction

Figure 1.3: Genome organization of DENV: Genomeorganization of DENV starting with 5'UTR along with three major structural genes (C, prM and E) while ending at the

3'UTR region in between there are seven nonstructural genes (NS1, NS2A, NS2B, NS3,

NS4A, NS4B and N55). Both structural as well as non structural genes play a vital role in replication of DENV(Guzman et al., 2010).

1.4 Replication and life cycle of DENV

In humans major targets for DENV replication are immune cells; during the early phase of DENV infection virus enters macrophages, monocytes and dendritc cells (DC).

After entry and replication DENV spreads to several organs including Liver, kidney and spleen (Upanan et al., 2008). While in mosquitoes initial sight of replication of DENV is the midgut region, later on DENV spreads to several organ(Mercado-Curiel et al., 2008;

Molina-Cruz et al., 2005; Salazar et al., 2007).

Adsorption studies revealed that DENV enters into the host cell via clathrin mediated endocytosis (Heinz et al., 2004). Next step involves the release of Nucleocapsid

(NC) in the cytoplasm, naked genome is the replicated and translated for production of progeny viral particles utilizing host machinery. Translation of DENV RNA results in formation of endoplasmic reticulum (ER) bound poly-protein which is later cleaved to form structural and non structural proteins(Cahour et al., 1992). After the cleavage of structural proteins the interaction between these proteins and genome results in encapsidation of DENV RNA, DENV C-RNA complex formation occurs in the membrane of ER, nevertheless this whole interaction is still unclear (Ma et al., 2004).

The process of virus assembly and release is still not clear but the process of virus

6

Chapter 1 Introduction

maturation depends on the activity of cellular enzyme furin, pre-membrane (prM) is cleaved to membrane (M) by furin which is the final step of virus maturation before release form the host cell(Stadler et al., 1997) as shown in Figure 1.2.

Figure 1.2: Replication cycle of DENV: Complete Following initial attachment the particle engages in further interactions and finally enters cells by receptor-mediated endocytosis. The viral RNA genome is released into the cytoplasm and translated at the rough ER, giving rise to a polyprotein that is cleaved into mature proteins. RNA replication occurs when negative-sense copy (–RNA) that serves as a template for the production of excess amounts of positive-sense progeny RNAs (+RNA). Assembly of

7

Chapter 1 Introduction

DENV particles initiates in close proximity to the ER, where core protein and viral RNA accumulate. After acquiring viral envelope virus buds out of cell(Alcaraz-Estrada et al.,

2010).

1.5 PATHOGENESIS

Pathogenesis of DENV starts at the site of mosquito bite, DENV enters into the

DC and macrophages where initial replication takes place (Marovich et al., 2001; Wu et al., 2000). Subsequently DENV migrate to the lymph nodes for further multiplication and spreading, from these lymph nodes DENV enter into the blood stream and spread throughout the body. DENV virus can replicate in several cells, however, DENV prefers immune cells like monocytes, DC and macrophages for replication (Halstead et al., 1977;

King et al., 1999). During a secondary DENV infection a high titer of IgM is produced which binds with DENVs and assists in engulfment by monocytes. Replication of DENV results in apoptosis of infected monocytes, this leads to stimulation of DCs and production of pro-inflammatory cytokines (Bosch et al., 2002; Espina et al., 2003;Palmer et al., 2005).

Diversity of DENV host cells indicates the involvement of several receptors for viral attachment and entry, recent studies resulted in identification of several host cells molecules such as GRP78/BiP, Hsp90, high-affinity laminin receptor, heat-shock protein

70 (Hsp70) and R67 which were responsible for DENV entry in the host cells(Chen et al., 1999; Chen et al., 1997b; Germi et al., 2002; Hilgard and Stockert, 2000;

Jindadamrongwech et al., 2004; Mendoza et al., 2002; Thepparit and Smith, 2004).

DENV also infects human granulocyte precursor cells (myeloid cells) via C-type lectin receptors (CLR). These receptors are C-type lectin domain family 5, member A (CLEC5,

8

Chapter 1 Introduction

MDL-1), intracellular adhesion molecule 3 (ICAM-3) and mannose receptor (MR)

(Fernandez-Garcia et al., 2009; Lozach et al., 2005; Miller et al., 2008; Navarro‐Sanchez et al., 2003; Tassaneetrithep et al., 2003).

Individuals infected with a serotype of DENV produce a lasting immunity however this immunity is specific for that serotype not for other serotypes. If an individual infected with one serotype is infected with another serotype this cross- reactivity can lead to DHF and DSS (Halstead, 2006; Sabin, 1952). Antibodies including

IgG produced during DENV infection in mother can reach fetus, afterward if a new serotype infects the new born this can lead to DHF and DSS.

Recent studies have suggested that pathogenicity is highly variable amongst all four serotypes and the level of virulence achieved is dependent on each individual serotype (Rico-Hesse et al., 1997). Epidemiological data suggests that Asian serotypes were able to produce higher virus titers in mosquitoes as well as in humans which contributed to increasing outbreaks (Cologna et al., 2005). Although vertical transmission is possible in mosquitoes however it is a rare occurrence, the highest rates of vertical transmission is seen in Aedesmediovittatus which acts as a major reservoir for

DENV(Freier and Rosen, 1988).

1.6 DENV IN VITRO CULTURING

Dengue virus can be grown in vitro in several cell lines originating from numerous animals and insects, these cell lines were established by extensive research on different cell lineages like myeloid-derived cells, hepatocytes, lymphocytes and kidney cells (Andrews et al., 1978; Kurane and Ennis, 1988; Kurane et al., 1984). DENV has been isolated from phagocytic cells (monocytes and macrophages),

9

Chapter 1 Introduction

polymorphonuclearleukocytes (PMNs) and leukocytes as these cells engage DENV upon infecting a new human host (Halstead et al., 1977; King et al., 1999;Scott et al., 1980).

Further research has revealed that dendritic cells (DC) along with macrophages having DC-SIGN and mannose receptorsarefavorable for DENV infection and replication

(Kyle et al., 2007). As skin is the initial site of entry of DENV after it bites a human host therefore langerhan cells of skin were also found to be permissive for DENV infection(Schmid et al., 2014).

1.7 ANIMAL MODELS

Animal model studies of DENV has been a challenge for researchers owing to limited replication in wild type (WT) mice and asymptomatic clinical disease in primates other than humans, earliest approaches involved intra-cerebral (IC) inoculation of DENV in suckling mice (Raut et al., 1996). Later adult mice having a normal immune system which were injected which resulted in neurological malfunctions and in severe cases paralysis was observed (Johnson and Roehrig, 1999).

AG129 mice lacking α/β and γ receptors were used for DENV replication by intravenous injections containing high doses of DENV (Johnson and

Roehrig, 1999). This development lead the way for future studies concerning DENV pathogenesis, inhibitor development and antibodies testing (Balsitis et al., 2010; Shresta et al., 2006). Mice and other non human primates lack clinical symptoms and disease manifestations still there are important for virus propagation as DENV can infect and replicate in multiple cells (Bente and Rico-Hesse, 2006; Kuruvilla et al., 2007).

1.8 RNA INTERFERENCE

10

Chapter 1 Introduction

RNA interference (RNAi) is an innate defense and regulatory process of eukaryotic cells for controlling or silencing of gene expression through small double stranded RNA (dsRNA) (Almeida and Allshire, 2005) as shown in Figure 1.4. Small interfering RNAs (siRNAs) are ∼21-22 bp in length with a 3' overhang of 2 nucleotide, this 3' overhang is responsible for RNAi based degradation of target mRNA for achieving gene silencing.

Figure 1.4: A typical siRNApathway:RNAi pathways usually start via small RNAs which can be small interfering RNA (siRNA) or microRNA (miRNA).siRNA pathway

11

Chapter 1 Introduction

start with the cleavage of double stranded RNA (dsRNA) by Dicer enzyme complex, these siRNAs get incorporated into Argonaute 2 (AGO2) and RNAi induced silencing complex (RISC). AGO2 cleaves the sense strand while active RISC uses the anti-sense strand to find its way to target mRNA sites for cleavage (Kim and Rossi, 2007).

1.9 INSILICO ANALYSIS

Molecular characterization of DENV and in depth phylogenetic analysis has resulted in four different serotypes (DENV1, 2, 3 and 4). These serotypes have differences at the genome level which is responsible for serotype diversity(Kuno et al.,

1998). Within serotypes several genotypes are present which result from geographical distribution and mutation amongst viral strains (Araújo et al., 2009; Rico-Hesse, 1990;

Twiddy et al., 2002; Villabona-Arenas and de Andrade Zanotto, 2011). DENV has a

RNA genome with a poor proof reading mechanism; the viral enzyme RNA-dependent

RNA polymerase has a high error rate.

Studies have revealed the fact that during one complete genome replication cycle one mutation occurs and collectively these mutations contribute in diversity amongst serotypes and genotypes (Holmes, 2003; Steinhauer et al., 1992). Mutation studies have shown that the largely deletion mutations are involve in evolution of DENV and emergence of several strains (Sall et al., 2010). The high mutation potential and genetic diversity in the DENV population has contributed to rapid evolution rate (Dunham and

Holmes, 2007; Villabona-Arenas and de Andrade Zanotto, 2011).

Epitope mapping is a laborious process which involves vast peptide screening as well as studying various aspects of immunogenic proteins via proteomics (Muzzi et al.,

2007). Wet lab experiments require large quantities of blood samples in addition factors

12

Chapter 1 Introduction

like low prevalence of specific epitopes, assortment of HLA alleles and cost of peptide synthesis makes epitope mapping an uphill task (Sette et al., 2001). To overcome these problems epitopes can be efficiently mapped by online Bioinformatics tools (Vázquez et al., 2002).

1.10 AIM AND OBJECTIVES

Current study was aimed at inhibiting DENV2 and DENV3 in mammalian cell line via RNA interference. A complete lay out of the study includes propagation of

DENV2 and DENV3 both mature and immature viruses. RNAi based inhibitory studies of siRNAs against both DENV2 and DENV3 serotypes. Inhibition of mature DENV2 grown on U-937 DC-SIGN cells. The second phase of study included isolation and propagation of Pakistani DENV2 isolate in suckling mice model.Insilico analysis of local gene sequences for prediction of future vaccine targets.

13

Chapter 2 Review of Literature

Chapter 2

LITERATURE REVIEW

Dengue fever was mentioned in Chinese medical encyclopedia dating back to 992 A.D.

Historical accounts from 18th century suggest the presence of a disease with symptoms resembling DF, epidemics were seen all across the globe from China to Americas.

Tropical and sub tropical regions having higher vector population had sporadic DF outbreaks (Gubler, 1998; Huang et al., 2000). A DF like disease was reported after

World war II in the region of Southeast Asia, patients were mostly children (Rigau-Pérez et al., 1998). Outbreaks were a common occurrence in 1950s, this led to documentation of outbreaks and in 1954 DHF outbreak in Manila was documented for the first time(Monath, 1994).

More than half (3.6 million) of the world's population is living in the tropical and sub-tropical regions of the world, owing to the presence of DENV vector (Aedesaegypti) these regions present a higher potential for DENV outbreaks (Ferreira, 2012; Gubler,

2011; Guzman et al., 2010). According to recent reports approximately 200 million case of DF occur each year, from these cases half million (500,000) progress to complications like DHF and DSS. Annual mortality rate is approximately 20,000 in different regions of the world, many cases are not reported which suggest that the number of cases might be higher (Gubler, 2002; Shepard et al., 2011).

Members of Flavivirus are present all across the planet and several of these viruses are associated with human diseases, some of the prevalent members include

Japanese encephalitis virus (JEV), (WNV), dengue virus (DENV), Tick-

23

Chapter 2 Review of Literature

borne encephalitis viruses (TBEV) and virus YFV (Vasilakis and Weaver,

2008). Phylogenetic analysis of viruses in Flavivirus genus revealed that all of these members are closely related based on vector interactions and antigenic properties (Kuno et al., 1998). The vector-borne viruses are further classified into mosquito and tick-borne viruses, three prominent tick-borne are virus, tick-borne encephalitis virus and virus. While mosquito-borne viruses are transmitted largely by Aedes and Culex genera and these mosquitoes are the main reason for global outbreaks of DENV, YFV and JEV (Kuno et al., 1998).

2.1 GENOMIC ORGANIZATION OF DENV

DENV belongs to Flaviviruseshaving a single stranded RNA genome which is approximately 11 kilo bases (Kb), the infectious positive sense RNA is directly translated into viral polyprotein(Boulton and Westaway, 1977; Rice et al., 1985). The diverse features is the presence of a solitary open reading frame (ORF) which encodes a poly- protein which is later cleaved to produce all ten structural as well as non structural proteins. Viral RNA has non coding regions (NCR) or untranslated terminal repeats

(UTR) at both ends, 5' UTR is capped whereas 3'UTR is deficient in a poly (A) tail

(Wengler et al., 1978) Table 2.1. 5' UTR assists in translation of viral RNA and viral replication, RNAi based approaches targeting this region was able to block both of these functions (Deas et al., 2005; Holden et al., 2006). The 3'UTR region is variant amongst

DENV and other and only a small stretch of nucleotides is conserved

(Markoff, 2003). 3'UTR has also shown to play role in viral RNA translation and replication by interacting with viral replicase enzymes (Chen et al., 1997a).

24

Chapter 2 Review of Literature

Structural region of DENV RNA genome is located near the 5'UTR which encodes three major structural proteins Capsid (C), precursor of matrix (prM) and

Envelope (E) whereas nonstructural (NS) region is the remaining portion of genome stretching till 3'UTR encoding all seven NS proteins (Rice 1985). The poly protein is produced from a single ORF (5'-C-prM/M-E-NS1-NS2A-NS2B-NS3-NS4A-NS4BNS5-

3’), large scale cleavage of this poly-protein results in the production of structural and NS proteins. All of these processes take place inside the membranes structures located in the cytoplasm of host cell, transportation of proteins is achieved through cellular signal sequences and if these structures are disrupted by external means can lead to inactivation of protein processing(Svitkin et al., 1984). Studies have demonstrated that several structural and nonstructural proteins are cleaved by host enzymes such as signal peptidase, furthermore viral serine protease is also responsible for cleavage of C and major NS proteins (von Heijne, 1984). For virus maturation host enzyme Furin protease cleaves precursor prM into mature M protein inside the Golgi network (Stadler et al.,

1997). Thus host as well as viral enzymes cleaves Flavivirus poly-protein into individual structural and NS proteins for formation of progeny viruses.

2.2 DENV PROTEINS

Amongst structural proteins E is considered as the most important proteins because E protein is involve in receptor binding with host cell receptors for attachment and fusion.

Host antibodies also interact with antigenic epitopes on E protein, consequently most vaccines are derived from E proteins to achieve maximum immunogenicity(Chambers et al., 1990). The M glycoprotein is also immunogenic to some extent however neutralizing antibodies are mostly produce against E protein (Kaufman et al., 1989).

25

Chapter 2 Review of Literature

Envelope is the largest structural protein which is involve in binding with host cell surface receptors and fusion for viral entry (Allison et al., 2001; Crill and Roehrig,

2001). DENV surface is relatively smooth owing to the E homodimers present all across virus surface (Kuhn et al., 2002). Studies relating to structure of DENV revealed that E protein has three domains (EDI, EDII and EDIII) (Rey et al., 1995). These domains have some important functions, ED1 works as a flexible hinge regions which assists in viral fusions.ED2 has two sections (Adams et al., 1995; Guirakhoo et al., 1989). EDIII plays a key role in DENV endocytosis by interacting with host cell receptors, EDIII is also interacts with heparin sulfate receptors of host cells that attachment is followed by fusion and entry (Chu et al., 2005; Huerta et al., 2008; Hung et al., 2004; Krishnan et al., 2007).

Studies have shown that E protein is a type II fusion protein which fuses with cellular endosome membrane via highly hydrophobic region in the EDII (Lescar et al., 2001; Rey et al., 1995).

Pre-membrane (prM/M) is the second structural protein having a size of

19kDa,prM is linked with capsid by a hydrophobic stretch. During DENV maturation prM is converted into matrix (M), the precursor prM contains N-linked glycosylation sites at the N terminal and six conserved cysteine residues which are responsible for disulfide bridging (Chambers et al., 1990; Nowak and Wengler, 1987). Both prM and E have transmembrane domains, these domains perform the role of retention signals and supports heterodimer formation (Lin and Wu, 2005)as shown in Table 2.2. DENV maturation involves cleavage of prM to M this leads to the viral budding via attachment with host cell membrane (Li et al., 2008). Heterodimerization of prM/E results in release of prM resulting in the E homodimers(Stiasny et al., 1996; Wengler, 1989).

26

Chapter 2 Review of Literature

Capsid protein is the smallest structural protein which is basic in nature having a size of 12kDa, C protein is cleaved from polyprotein by NS3/NS2B DENV proteases and host cell signalase and consequently released in cytoplasm (Amberg and Rice, 1999;

Lobigs, 1993). Studies have shown that homodimer form of C protein and DENV nascent

RNA fuse to form nucleocapsid which buds off into the ER Lumen to initiate virus formation by interacting with prM and E proteins (Lindenbach and Rice, 2001; Ma et al.,

2004;Wang et al., 2004) as shown in Table 2.2. C protein is essential for DENV assembly, the importance of C protein becomes obvious with the production of sub-viral particles lacking DENV genome as well as C protein(Ferlenghi et al., 2001).

DENV has seven non structuralproteins; the precise role of these NS protein is still not properly understood. Recent studies suggest that NS protein are responsible for viral replication and encapsidation, moreover these NS are also involve in evading the host antiviral responses (MACKENZIE et al., 1996; Muñoz-Jordán et al., 2003).

NS1 is a 40-46 kDa protein which monomeric and changes to dimeric after few minutes (Falgout et al., 1989).NS1 is highly abundant in cells it's found on cell surface and is also released from the cell. NS1 plays a key role in two major processes, NS1 in association with DENV RNA and human heterogenous nuclear ribo-nucleoprotein

(hnRNPC1/C2) produces nascent mRNA (Noisakran et al., 2008). NS1 is an important mediator in RNA as well as viral replication (Mackenzie et al., 1998).

The precise role of NS2A is not clear nevertheless it is involve in DENV replication. A recent study showed that mutations in NS2A caused hindrance in viral assembly and subsequent replication of DEN (Xie et al., 2013). During DENV infection

27

Chapter 2 Review of Literature

NS2A has shown to inhibit interferon (IFN) signaling assisting DENV to avoid host immune system (Simmons et al., 2010).

NS3 has multiple functions like helicase, protease and genome capping. DENV mRNA is translated into a poly protein; this polyprotein is processed by NS3 protease domain. NS2B act as a co-factor for NS3 and assists NS3 in its protease activity (Erbel et al., 2006). Recent studies have suggested that NS2B/NS3 complex can activate caspases which in turn can lead to apoptosis of host cell (Shafee and AbuBakar, 2003). NS3 has a helicase activity owing to C-terminal amino acids (170-619), this portion of NS3 protein has also shown RTPase and NTPase/helicase activity. The helicase functions by splitting the new DENV RNA from parent template in addition to that the helicase activity results in the unwinding of secondary structures at 3' UTR for starting replication (Gorbalenya et al., 1989).The interaction of hypophosporylated form of DENV NS5 and NS3results in the activation of both RTPase and NTPase. RNA capping is also via involvement of 5' triphosphatase (RTPase) (Bartelma and Padmanabhan, 2002; Yon et al., 2005).

NS4A is one of the smallest DENV protein, owing to its hydrophobia nature it is mostly present in association with host membranes (Miller et al., 2007). Main role of

NS4A is the proper organization of cellular membranes for allowing replication of DENV genome.

NS4B is a hydrophobic protein having a size of 27kDa present throughout the cytoplasm of infected cell; however it is initially found in peri-nuclear regions. NS4B has shown to interact with NS3 helicase region which is indicative of the fact that NS4B is an important component of viral replication (Miller et al., 2006). NS4B has two major roles in DENV RNA replication one of those is assisting in the detachment of NS3 helicase

28

Chapter 2 Review of Literature

from RNA, whereas the second role of NS3 is that it clutches both strands and keeps them separated during RNA replication (Umareddy et al., 2006). NS4B in collaboration with NS4A and NS2A contribute in resisting host IFN activity(Muñoz-Jordán et al.,

2003).

NS5 is the largest of all NS DENV proteins with a size of 104 kDa, detail analysis revealed that NS5 has a conserve sequence amongst DENV serotypes and even some other member of Flaviridae family (Yap et al., 2007). NS5 can be divided into two parts one is involve in capping at the 5' end of DENV genome while the C terminal of NS5 is a

RNA dependent, RNA polymerase that produces intermediate double stranded RNA finally resulting in the production of positive sense RNA for progeny viral particles

(Egloff et al., 2002; Zhou et al., 2007).

29

Chapter 2 Review of Literature

Table 2.1: DENV RNA elements and there functions.

RNA ELEMENTS FUNCTIONS

5' UTR Non canonical translation and viral RNA synthesis

cHP Translation initiation codon selection and viral

viability

5'/3' UAR Viral RNA cyclization and viral viability

5'/3' CS Viral RNA cyclization and viral RNA synthesis

3' UTR Cap dependent non canonical translation and viral

RNA synthesis

VR Translation and viral RNA synthesis

DB1/DB2 Translation and viral RNA synthesis

3'SL Translation and viral RNA synthesis Table

2.2: DENV proteins and there functions.

30

Chapter 2 Review of Literature

VIRAL PROTEIN FUNCTION

C Viral RNA packaging

prM Prevention of premature fusion

E Receptor binding and fusion

NS1 Signal transduction

NS2B NS3 serine protease cofactor

NS3 Helicase, NTPase, 5' triphosphatase and serine protease

NS4B Inhibition of IFN signal transductin

NS5 RDRP and methyltransferase

31

Chapter 2 Review of Literature

2.3 OVER VIEW OF DENGUE VIRUS

Dengue virus infection is the most prevalent mosquito borne viral infection.

DENV belongs to family Flaviviridae having four serotypes i.e. DEN-1, DEN-2, DEN-3 and DEN-4. In humans DENV causes dengue fever (DF) which can develop into severe complications like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS)

(Kyle and Harris, 2008). Reemergence of DENV in recent years has developed into a grave health risk in tropical and subtropical regions of the world (Barrera et al., 2002; da

Glória Teixeira et al., 2002;Sukri et al., 2003). Flaviviruses in general encode for three structural proteins i.e. capsid (C), pre-membrane (prM) and envelope (E) proteins and seven nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5).

Structural proteins develop into a viral particle while nonstructural proteins play role in viral replication and assembly (Kümmerer and Rice, 2002; Liu et al., 2003).

Anti DENV inhibitors are designed keeping in view potential host and viral targets where various mechanisms can be blocked to control disease progression like enzymatic interactions, viral replication, host and viral receptors interaction, etc.

Antiviral agents that can reduce viral titer many folds can decline high mortality rates associated with DHF/DSS (Noble et al., 2010; Vaughn et al., 2000). Strategies to curb

DENV usually include targeting viral enzymes and replication mechanism, while host machinery can also be targeted with the added advantage of reduced chances for development of viral drug resistance.

Current treatment options include hemorrhagic condition management and fluid therapy with no targeted drugs for viral clearance. As development of effective vaccine has still not been achieved, development of an effective therapeutic agent against DENV

32

Chapter 2 Review of Literature

is vital to bring down annual DENV related mortality rate, a number which usually exceeds the reported one (Whitehead et al., 2007). DF annual epidemiological estimates lack accuracy due to multiple factors, which include large number of unreported cases, absence of proper diagnostic procedures such as determination of serotype and in a few cases diagnostic errors(Farrar, 2008; Range, 2008).

Research in recent years can and has led to the development/identification of possible therapeutic agents for DENV, also advances in drug delivery mechanisms can target the precise area with high efficacy and negligible chances of development of side effects (Pollock et al., 2010). Host factors play a key role in development of DF, a recent study discovered 42 human factors that contribute appreciably in the development of

DENV infection (Sessions et al., 2009). Thus a detail study of these factors can open many avenues for development of anti-dengue therapeutics.

Vaccine approaches against DENV date back to 1929, when for the first time formalin phenol inactivated vaccine was developed. Although this vaccine was unsuccessful still it marks the beginning of vaccine strategies against DENV(Simmons et al., 1931). Later attempts involving live attenuated vaccine were also stopped over growing concerns regarding patient safety (Hotta, 1952; Wisseman et al., 1963). Since

DENV is an enveloped virus, antibodies are produced mostly in response to the highly immunogenic envelope protein (Brinton et al., 1998). Vaccinia vector based vaccine having truncated envelope gene provided complete protection to a DENV challenged monkey (Men et al., 2000). Adenovirus vector vaccine has also been very effective, enhancing immune response in vitro against all four DENV serotypes (Raviprakash et al.,

2008).

33

Chapter 2 Review of Literature

2.4 DENV SEROTYPES AND GENOTYPES

Although DENV has been classified into four major serotypes however each serotype can have several genotypes depending on geographical distribution and place where that strain originated (Rico-Hesse, 1990). DEV1 has five genotypes and members of genotype I are from China, East Africa and parts of Southeast Asia. Genotype II is based on DENV1 from Thailand whereas DENV1 isolated from Malaysia are grouped into genotype III (Goncalvez et al., 2002). DENV1 isolated from Australia and Pacific

Island were placed in genotype IV and lastly genotype V contained DENV1 from West

Africa, South America and some countries from Asia (Wang et al., 2000).

DENV2 has five genotypes originating from various regions of the world, genotype I is further divided into two sub groups Asia I which has Malaysian strain of

DENV2 while Asia II contains DENV2 isolates from Sri Lanka, Taiwan, Philippines and

Vietnam. Genotype II contains strains for several countries, isolates of DENV2 from

Pacific Islands, Indian subcontinent, American and some Caribbean countries make up genotype III. The genotype IV has DENV2 isolates from Asian countries including

Thailand, Vietnam and Americas. Final genotype V has strains from Southeast Asia and parts of West Africa (Lewis et al., 1993).

DENV3 is classified into five genotypes among which genotype I contains strains from Philippines, Indonesia, Malaysia and South Pacific islands. Genotype II is based on strains from Thailand and genotype III is a group of isolates from Samoa, Africa, Sri

Lanka and India. Isolates from Tahiti and Puerto Rico make up genotype IV moreover strains from several Asian countries including China, Malaysia and Philippines are grouped into genotype V (Lanciotti et al., 1994; Wittke et al., 2002). DENV 4 has 4

34

Chapter 2 Review of Literature

genotypes and isolates from Japan, Thailand, Sri Lanka and Philippines are members of genotype I. Strain of DENV2 isolated from Tahiti, Americas, Malaysia and Indonesia are grouped into genotype II while genotype III contains strains from Thailand. Lastly genotype IV contains strains of DENV 4 isolated from wild animals (Lanciotti et al.,

1997).

2.5 PATHOGENESIS

Pathogenesis of DENV is inter-dependent on several factors which can modulate

DF to its complicated manifestations known as DHF/DSS. Worldwide DENV is the leading vector borne viral infection associated with highest mortality rates in children, both DHF/DSS are responsible for this high mortality rate (Anders et al., 2011).

Important factors in DENV enhancing severity of DF are explained below.

2.5.1 High virulence

Studies have shown that several DENV strains have a tendency for high virulence in comparison with normal DENV strains. This phenomena was initially observed by

Rosen and Gubler in 1970s when it was examined that some of the DENV strains caused limited number of DHF/DSS cases while SAME DENV had a higher ratio of DHF/DSS cases (Gubler et al., 1978). A similar case was seen in 1080s when a Southeast Asian strain of DENV2 was introduced in Cuba which lead to an outbreak of DHF in South

America (Guzman et al., 1995). With the onset of genomic sequencing phylogenetic analysis revealed that some strains of DENV were associated with severe DF infections and higher mortality rates (Messer et al., 2003).

2.5.2 Host Innate Immunity

35

Chapter 2 Review of Literature

Humans have a robust immune system and whenever any foreign antigen enters into the body immune response neutralizes the antigen. In humans one of the important component of innate immunity is complement system having several active proteins components.Studies have shown that patients with DSS have increased levels of complement proteins (C3a and C5a) in the plasma (Churdboonchart et al., 1983; Shaio et al., 1992). Complement activation is via antibodies which interact with DENV NS1 protein, this NS1 is present on the surfaces as well as in fluids after being released from

DENV infected cells (Avirutnan et al., 2006; Kurosu et al., 2007). Two complement proteins C5b-C9 which work as a complex are associated with progression of DF into complications like DHF/DSS.

2.5.3 Antibody Dependent Autoimmune Responses in DF

During DF human body mounts an immune response to neutralize DENV, this neutralization takes place by the help of antibodies directed against several viral antigens.

Most of these antibodies are produced against E protein which is the largest DENV structural protein, however a small portion of DENV E protein has a sequence resemblance with numerous host factors including prothrombin, plasminogen and tissue plasminogen factor (Huang et al., 1997; Markoff et al., 1991). Platelets are also a target for cross reactive antibodies which are initially generated against DENV NS1 protein, these cross-reacting antibodies are potent inhibitors of platelet aggregation and can cause severe thrombocytopenia (Chen et al., 2009; Lin et al., 2008a). Amongst several NS1 cross-reactive antibodies the most important antibody is IgM which can activate pro- apoptotic caspase in endothelial cells and this antibody can trigger secretion of inflammatory cytokines i.e. MCP-1, IL-6 and IL-8(Lin et al., 2005).

36

Chapter 2 Review of Literature

2.5.4 Cellular Autoimmunity in DHF/DSS

T cells are crucial part of cellular immunity against DENV as they can clear infected cells and limit DENV replication. Research in T cell studies have demonstrated that although CD8 cells are able to clear DENV however these calls have a potential to cause cross reactivity(An et al., 2004). Highly cross reactive CD8 cells are associated with production of inflammatory cytokines IL-13, TNFα and IFNγ which can cause severe inflammation in human host. These hyper active CD8 cells are short live and after producing pro-inflammatory cytokines these cells die by apoptosis (Dong et al., 2007;

Mongkolsapaya et al., 2006). Studies have shown that these hyper active CD 8 cells are responsible for mass production of pro-inflammatory cytokines however the ability of these cells to produce cytotoxic granules is lost (Hober et al., 1993). Serum analysis of

DHF/DSS patients have revealed soaring levels of pro-inflammatory cytokines (IL-1β,

IL-6, IL-8, TNFα and IFNγ), owing to immuno-pathogenic CD8 cells (Hober et al.,

1993). Initially research on immuno-pathogenesis of DENV revealed that cross reactive

T cells are generated against NS proteins, later on studies conducted on mice models demonstrated that hyper active CD8 cells are produced against both structural (prM and

E) as well as NS (NS1, NS2a and NS3) (Beaumier and Rothman, 2009).

2.5.5 Antibody Associated with Enhancement of DF

Antibodies are important component of human immune response for neutralizing

DENV however some antibodies can further complicate the situation by enhancing DF

(Guzmán et al., 1990). Higher rates of infant mortality have been associated with

DHF/DSS owing to the presence of maternal antibodieswhich have lost ability to neutralize DENV till the age of six months however these antibodies keep reacting with

37

Chapter 2 Review of Literature

DENV until one year (Chau et al., 2009). These antibodies lacking neutralizing potential contribute in ADE. In children older than one year sometimes a prior DENV infection can result in production of non-neutralizing antibodies which can cross react with a subsequent serotype of DENV and lead to DHF/DSS (Halstead et al., 1973; Halstead et al., 1970).

For ADE weakly neutralizing antibodies require immune complex for proper attachment with phagocytes (Halstead and O'rourke, 1977). Fc receptors (FcR) have also been extensively studied for their involvement in ADE, two FcR (FcγRI and FcγRII) were identified for playing crucial roles in development of ADE whereas the significance of FcγRIII in ADE is still not properly understood (Littaua et al., 1990; Unkeless, 1989).

A study on AG129 mice having modified FcRs revealed low virus replication and lack of severe disease (Balsitis et al., 2010). DENV titers go up rapidly during ADE owing to the enhancement of infection in host cells, one of the factor which contributes to elevated rate of virus replication is the conversion of immature virus into viral receptors via

FcR(Balsitis et al., 2010; Libraty et al., 2002).Another possible way of ADE involvement in DHF/DSS is via down regulation of cellular nitric oxide, this results in the inhibition of

STAT1 and IRF1 which in turn limit the innate immunity (Wang et al., 2006).

According to most studies FcR were responsible for ADE however later research revealed that cells lacking FcR were also involved. Research conducted by Halstead and his fellow researcher demonstrated the crucial role of IgG in developing ADE

(Chareonsirisuthigul et al., 2007).

2.6 Viral targets for DENV inhibitors

38

Chapter 2 Review of Literature

Dengue viral proteins play an important role in the pathogenesis and propagation of the virus, these viral proteins are mostly enzymes, that are absent in the host cells thus minimizing the risks associated with inhibitors interfering in host cell mechanisms..

Therefore these proteins can be selectively targeted by inhibitors for viral control and clearance.

2.6.1 Structural proteins’ based inhibition of DENV (Receptor mediated entry inhibition)

DENV envelope protein (E) belongs to class II viral proteins engaged in fusion with host cellular receptors (Harrison, 2005). E protein regulates fusion with numerous host receptors which include glucose regulated protein 78 (GRP78), DC-SIGN, L-SIGN, laminin receptors, mannose receptors and ICAM-3 grabbing non-integrin receptors

(Jindadamrongwech et al., 2004; Miller et al., 2008; Navarro‐Sanchez et al., 2003;

Tassaneetrithep et al., 2003; Thepparit and Smith, 2004). Thus these host receptors can be targeted for inhibiting virus/host interactions leading to infection. Since E protein mediates membrane fusion with host by regulating low pH-induced conformational changes and organizational rearrangements at the surface of virus, this membrane fusion can also be targeted for development of an anti-viral agent.

Structurally DENV E protein has three domains i.e. I, II and III, where domain

III has shown potential for blocking viral entry into the host cell by disruption of viral fusion (Barbas et al., 1997; Liao and Kielian, 2005; Modis et al., 2004). DC-SIGN receptors were blocked by carbohydrates likeplantlectins (HHA) binding agents reducing

DENV infection but due to their specificity to DC-SIGN receptors these agents are inefficient to be used for cells lacking these receptors (Alen et al., 2009).

39

Chapter 2 Review of Literature

2.6.2 Nucleoside based inhibition of DENV

Nucleosides enter the cell via two mechanisms: passive diffusion or through a nucleoside transporter (Pastor-Anglada et al., 2005).Variations have been observed in responses of different cells lines against DENV challenge due to varying levels of nucleoside and nucleotide kinases, therefore for a nucleoside inhibitor approach, multiple clinically relevant cell lines should be evaluated (Leary et al., 2002). A 2´C methyl deaza- nucleotide inhibitor showed wide range of anti-flaviviral activity targeting specifically RNA dependant RNA polymerase (NS5) (RdRp) of DENV, HCV and other members of Flaviviridae. A recent study found this inhibitor highly effective against HCV-infected chimpanzee model (Carroll et al., 2009; Migliaccio et al., 2003).

DENV was also effectively inhibited by an adenosine analog (NITD008) both in vivo and in vitro studies, this analogue contain two key substitutions. A carbon was substituted at N-7 position of purine and acetylene was added at 2’ position of ribose

(Yin et al., 2009). In a recent study a has shown promising prospects to inhibit dengue virus NS5 protein. There are also certain other guanosinenucleoside analogues including acyclovir and EICAR that effectively compete with GTP binding to DENV NS5 protein (Benarroch et al., 2004).

2.6.3 Methyltransferases inhibition of DENV

Genome of Flaviviruses has a 5´cap and this capping is achieved by a series of enzyme regulated reactions. The 5´end of viral RNA is hydrolyzed by NS3 tri- phosphatase from tri-phosphate to di-phosphate, while a guanylyl-transferase transfers

GMP to 5´-diphosphate. NS5 methylates N-7 position of guanine, and 3´OH position of first nucleotide ribose that prepares the 5´ cap. DENV serotype 2 deficient in 5´ cap

40

Chapter 2 Review of Literature

methylation has shown inhibition of replication potential (Cleaves and Dubin, 1979;

Egloff et al., 2002; Ray et al., 2006; Wengler and Wengler, 1981; Wengler and Wengler,

1993).

In another approach seventeen mutations were introduced in amino acids of methyltransferase domain targeting specifically amino acids that were directly involved in s-adenosyl-I-methionine binding, or vital amino acids for proper confirmation of NS5 and charged amino acids involved in various interactions (Podvinec et al., 2010). In one of the studies, 10 novel inhibitors of NS5 methyltransferase were identified that interacted with NS5 in a similar fashion. Computer based molecular docking approach was applied to 5 million commercially available compounds in order to screen for inhibitors of methyltransferase, out of which 263 compounds were selected for further testing and it was observed that 10 compounds were effective for inhibition of methyltransferase(Lim et al., 2011). S-adenosyl-homocysteine (SAH) is a chemical derivative that binds to methyltransferase of dengue virus and selectively inhibits its replication, this binding is not related to human enzymes. Dengue virus methyltransferase crystalline structure exhibited that its N6-substituent bound in the cavity caused conformational changes in residues lining the pocket. Therefore, this study was vital since it proved that a virus specific methyltransferase inhibitor could be prepared to inhibit DENV replication (Luzhkov et al., 2007).

Combining computational and experimental screening techniques a new novel inhibitor for dengue methyltransferase has been identified with a previously unknown scaffold. Computer based approaches such as 2D similarity searching, pharmacophore filtering and docking were used to identify compounds that inhibited DENV

41

Chapter 2 Review of Literature

methyltransferase and 15 top hit compounds were selected and were further tested on recombinant methyltransferase. Study resulted in discovery of novel inhibitor for dengue virus that had an IC (50) value of 60 microM(Kroschewski et al., 2008).

2.6.4 Protease (NS2B/NS3) inhibition of DENV

Proteases (host and viral) play a vital role in the viral poly-protein processing.

During recent years RNA virus protease domain has been the target for many antiviral therapies. Protease inhibitors against HCV are in clinical trials and since DENV belongs to the same family, these approaches can therefore be utilized for development of therapeutic agents against DENV (Soriano et al., 2008).

DENV serine protease belongs to Trypsin superfamily having a catalytic triad

(His51, Asp75, Ser135) with more than 50% homology amongst members of genus

Flavivirus(Bazan and Fletterick, 1989; Valle and Falgout, 1998). Serine protease of

DENV has optimal activity at a high pH (pH 9) (Leung et al., 2001) and is resistant to available potent inhibitors of serine proteases. Many factors contribute to the ineffectiveness of protease inhibitors for DENV: the most prominent being weak binding of inhibitor with enzyme, where only aprotinin was found to be effectively inhibit DENV protease by blocking the active site (Ekonomiuk et al., 2009; Mueller et al., 2007;

Tomlinson et al., 2009; Yang et al., 2011).

In a recent study EUDOC docking program based library of small molecules was generated. These compounds exhibited binding to P1 pocket and catalytic site of DENV2

NS3 protease domain. Most favorable compounds obtained through docking were later used for in vitro inhibition of DENV2, these compounds showed high solubility and

42

Chapter 2 Review of Literature

inhibition of DENV2 in cell culture experiments. Final results showed that 2 compounds were most promising for inhibition of protease inhibition (Yin et al., 2006).

BP2109 is another dengue virus protease inhibitor that is associated with viral replication and viral RNA synthesis inhibition (Tomlinson et al., 2009). In another approach different synthesized protease inhibitors, inhibited NS3 protease of dengue virus (Wang et al., 2011). Computer based identified of inhibitors for DENV have also been evaluated for DENV2 protease inhibition. Two of these potential compounds further inhibited viral replication in cell culture experiments (Mueller et al., 2008).

2.6.5 Host enzymes based inhibition of DENV

DENV relies on host enzymatic machinery for its replication and many host factors assist in viral replication. Since host enzymes are vital for various cellular mechanisms therefore a cautious approach is required for minimizing drug induced toxic effects and means for their delivery.

Different inhibitors have been evaluated against host enzymes for dengue virus inhibition. Dengue virus requires host nucleosides for replication and these nucleosides are incorporated into new virus progeny. In a recent study a novel antiviral compound

(NITD-982) has been identified that can inhibit host dihydroorotate dehydrogenase

(DHODH), an enzyme required for pyrimidine biosynthesis. DENV was inhibited via diminishing intracellular pyrimidine pool, a major drawback of this anti-dengue was lack of effectiveness in vivo resulting from exogenous uptake of pyrimidine from diet(Aleshin et al., 2007).

2.6.6 Kinases based inhibition of DENV

43

Chapter 2 Review of Literature

Host kinases contribute in DENV replication by engaging in viral assembly and release from host cell. All four serotypes can be targeted via c-Src kinase inhibitors. In a study conducted in 2007 DENV assembly was blocked by dasatinib c-Src kinase inhibitor. c-Src, AZD0530 and other members of c-Src kinase inhibitors have also proven to be useful and safe anti-viral agents (Chu and Yang, 2007).

A study involving West Nile Virus (WNV) showed that c-Yes a member of Src kinase family was involved in virus maturation as the level of cellular Src family kinase c-yes was up-regulated when infected with WNV. Src Family Kinases (SFK) inhibitor pp2 reduced viral load up to 2-4 log in WNV infected cell lines, thus demonstrating that

Src family kinases have a pivotal role in viral replication and assembly inside the host.

RNA interference mediated down regulation of c-yes also resulted in decline in WNV titers suggesting that SFK are important host enzymes utilized by virus for its replication

(Hirsch et al., 2005).

2.6.7 Glucosidase based inhibition of DENV

Glucosidases are a group of enzymes that catalyzeglucoside hydrolysis, folding and glycosylation of various DENV structural and nonstructural proteins (E, prM and

NS1). In order to perform these functions DENV requires host glucosidase. Inhibition of host endoplasmic reticulum -glucosidase prevented folding of Pre-membrane and

Envelope glycoprotein of DENV (Hirsch et al., 2005). Such glucosidases can be potential targets as therapeutic against DENV. Deoxynojirimycin (DNJ) is an effective inhibitor of

Endoplasmic reticulum (ER) -glucosidase that inhibits posttranslational modification in a dose dependent manner. Castanospermin is an -glucosidase inhibitor that potentially

44

Chapter 2 Review of Literature

disrupts the folding of the structural proteins prM and E of all the four serotypes of

DENV (Whitby et al., 2005; Wu et al., 2002).

Iminocyclitol compounds have been tested effectively as antiviral compounds that affect the endoplasmic-reticular glucosidase mediated morphogenesis of DENV. These compounds specifically target budding viruses and show limited toxicity, these modifications can later be applied for development of anti-viral agents that can target multiple viruses (Gu et al., 2007).

2.6.8 Cytokines and receptors based inhibition of DENV

Immune system is involved in the progression of DF to DHF and DSS. Cytokine and chemokine including TNFα, IL-8, MMP-9 and VEGF/VEGF receptor have shown significant involvement in development of complications that lead to DHF and DSS

(Luplerdlop et al., 2006; Shresta et al., 2006; Souza et al., 2009; Srikiatkhachorn et al.,

2007). Recent studies attribute platelet-activating factor (PAF) with DENV pathogenesis.

Platelet-activating factor receptor (PAFR) has an important role in DENV pathogenesis and in a study it was observed that mice lacking PAFR showed decreased thrombocytopenia, hem-concentration, low levels of cytokines and a longer duration for manifestation of symptoms (Bai et al., 2007).

For limiting DENV pathogenesis regulatory mechanisms are required. In mouse models, antibodies produced against these cytokines provided protection but further research is required for their possible use in humans. Receptor mediated entry inhibition can also play a pivotal role in the inhibition of dengue virus.

2.6.9 Computational design of peptide inhibitors against DENV

45

Chapter 2 Review of Literature

Viral structural proteins mediate viral entry into the host cell via fusing with host cell receptors. Inhibitors blocking this fusion process can retard progression of infection.

In recent years a few inhibitors were designed and evaluated against beta sheet envelope

(E) proteins and cell attachment of DENV. These inhibitor peptides have shown promising results to inhibit DENV (Hrobowski et al., 2005; Rees et al., 2008). However, crystal structures of E protein both pre-fusion and post fusion were not exploited in most of the approaches for development of Flavivirus E protein directed inhibitors (Heinz et al., 1991; Kanai et al., 2006;Modis et al., 2003).

In a recent study inhibitor peptides were designed using residue-specific all-atom probability discriminatory function (RAPDF). X-ray diffraction structure of DENV-2 envelope protein served as a template for making mutant structures yielding mutant peptides (Zhang et al., 2004b). Residual side chains were substituted in all peptides by backbone-dependent side chain rotamer library and linear repulsive steric energy term provided by SCWRL version 3.0 (Bower et al., 1997). These peptides were successful in inhibiting DENV infection up to a certain extent.

2.7 Inhibition of DENV by siRNA

The production of siRNAs in eukaryotic cells occur by cleavage of long dsRNA sequences by endonuclease Dicer which is a complex protein having TAR-RNA binding protein (TRBP) (Zhang et al., 2004a). The next step involves the transfer of siRNA from

Dicer to RNA-induced silencing complex (RISC), RISC contains Argonaute 2 (Ago) protein having the potential of cleaving target mRNA strand. Ago are vital for proper gene silencing and in humans Ago-2 perform this cleavage (Liu et al., 2004; Meister et al., 2004).

46

Chapter 2 Review of Literature

Thermodynamic properties of siRNA targeting specific mRNA are vital for selection and attachment with RISC, mRNA molecule complementary to guiding RNA is cleaved by Ago-2 (Khvorova et al., 2003; Schwarz et al., 2003). One other class of RNAi is represented by 60-70 base pair hairpin shaped MicroRNAs (miRNAs) produced from precursor pri-miRNAs within the nucleus.

Production of miRNAs is achieved by complex known as Drosha-DGCR8 (Han et al., 2004; Lee et al., 2003). RNAse III Dicer excises the loop and consequently one strand is presented to RISC, this whole process takes place in the cytoplasm of the cell.

The miRNA shares a 3' UTRcomplementarity with target mRNA which is finally degraded for gene silencing (Bagga et al., 2005).

RNAi dependent specific RNA degradation involves double stranded RNAs

(dsRNA) which are abundantly present in the cytoplasm of eukaryotic cells (Elbashir et al., 2001; Fire et al., 1998; Montgomery et al., 1998). Gene silencing is achieved by the formation of RNA induced silencing complexes (siRNA) in association with various cellular factors (Hammond et al., 2000;Meister and Tuschl, 2004; Mello and Conte,

2004). Studies suggest that DENV could also be effectively inhibited by RNAi(Joost

Haasnoot et al., 2003; Sanchez-Vargas et al., 2004). It has also been suggested that mosquito’s continual infection without any pathogenic observations can attribute to

RNAi involvement (Sánchez-Vargas et al., 2009). Additionally, inside the host cytoplasm during the un-coating and replication processes, ssRNA viral genomes can be targeted by

RNAi(Uchil and Satchidanandam, 2003).

In a recent study siRNA produced against DENV glycoprotein gene precursors inhibited DENV replication and resulted in increased cell survival rate (Wu et al., 2010).

47

Chapter 2 Review of Literature

In Another study an engineered Sindbis virus containing 290 bp region of DENV serotype 2 prMregion produced resistance against DENV type 2 challenge. dsRNA were designed for DENV serotype 1 in a different study achieving reduction in viral load but it was less efficient as compared to Sindbis-induced silencing (Adelman et al., 2001;

Caplen et al., 2002). A more detailed analysis is required for the development of potential siRNA based inhibitors of dengue virus with multiple targets directed against more than one serotype.

2.8 VACCINE DEVELOPMENT FOR DENV

Development of vaccine against DENV is still a challenge for scientists across the globe. Presence of multiple serotypes pose a major problem as protection against a particular serotype can lead to a more severe condition in the form of DHF/DSS on co- infection with a different serotype due to phenomena known as antibody dependent enhancement (ADE) (Boonnak et al., 2008; Whitehead et al., 2003). A possible solution to this problem is development of a tetravalent vaccine which can provide protection against all four serotypes (DENV1-DENV4) (Konishi et al., 2006; Raviprakash et al.,

2008;Simmons et al., 1931). Historically efforts regarding development of DENV vaccine date back to early 20th century but these approaches failed to provide a suitable vaccine and in 1929 Formalin inactivated virus approach did not satisfy safety concerns

(Guirakhoo et al., 2004; Hotta, 1952; Wisseman et al., 1963).

Recent advances in molecular virology and immunology have opened new avenues in the field of vaccine development, latest approaches include sub unit vaccines,

DNA vaccines, chimeric vaccines and viral vector based vaccines.

2.8.1 Chimeric Vaccine

48

Chapter 2 Review of Literature

Chimeric vaccines contain fusion of two viral proteins resulting in high levels of protection. In a recent study a chimeric vaccine containing Yellow fever (YF) and DENV was prepared, Envelope (E) and prM region of YF were replaced by wild type (WT)

DENV representing all four serotypes. Chimeric vaccine viruses were electroporated into

Vero cells for production of chimeric viruses, 92% of monkeys survived when challenged after vaccination with chimeric YF/DENV vaccine (Trent et al., 2010).

Vaccines obtained from Vero cells contain higher amounts of cellular proteins,

DNA and cell debris, therefore a stringent process is required for purification of this formulation. Human diploid cell lines provide a better alternative with comparatively low levels of cellular debris (Raviprakash et al., 2008).

2.8.2 DNA vaccine

DNA vaccines provide added advantage of intracellular antigen processing and are safe as compared to live attenuated virus vaccines. Few concerns regarding DNA vaccines are; poor intake resulting in reduced antigen expression and weak immune response to incorporated structural proteins of virus. Alternatively addition of adjuvant may enhance immunity but is associated with increased cost related to manufacturing. In a recent study DNA vaccine in combination with recombinant viral vector vaccine was developed to tackle issues involving immunogenicity and gene delivery (Chen et al.,

2007b).

DENV serotype1 Envelope (E) and prM genes were expressed in Venezuelan

Equine Encephalitis (VEE) virus vector comprising of two doses of naked DNA with a third dose of VEE dengue particles, induced complete protection in macaques

(Ramanathan et al., 2009). Envelope conserved domain (Domain III) expressed in

49

Chapter 2 Review of Literature

mammalian expression vector has provided immunity against all the four serotypes in mice (McArthur et al., 2008).

Monkeys and mice have also been subjected to numerous DENV DNA based vaccines in an effort to reach a suitable candidate vaccine for humans (De Paula et al.,

2008; Konishi et al., 2000). Drawbacks of DNA vaccines are weak immune responses and high cost burdens and these factors demise the chances for the production of cost effective and potent DNA vaccine in near future (Wang et al., 1998; Wang et al., 2001).

2.8.3Live attenuated vaccines

Live attenuated vaccines (LAV) have been successful against Yellow fever virus

(YFV) and Japanese Encephalitis (JE) Virus. LAVs provide long term immunity and are cost effective as compared to DNA and Chimeric vaccines. Among other advantages,

LAV activate both types of immune responses i.e.humoral and cell mediated for a long period of time thus making LAVs ideal candidates for protection against DENV

(Edelman, 2007; Innis and Eckels, 2003).

Efforts for development of LAV for DENV have encountered a few problems from the beginning like low levels of immune activation and side effects that hampered the progress in development. For more than the last two decades monovalent LAV candidates have been propagated on primary and diploid cell cultures and later evaluated in humans to reach a final effective low cost vaccine (Edelman et al., 2003; Sun et al.,

2003).

A final break through was achieved by Halstead and Marchette in 2003 by multiple passaging of monovalent DENV LAV on primary dog kidney cell line (Halstead and Marchette, 2003). Monovalent DENV LAVs obtained from several passages on dog

50

Chapter 2 Review of Literature

kidney cell line were combined to make a tetravalent vaccine which was tested in phase I and phase II trials in adults and children from Thailand and America which was effective and provided protection. However certain unreported complications occurred in phase III trials which ultimately led to the suspension of this DENV LAV (Edelman et al., 2003;

SABCHAREON et al., 2004;Sabchareon et al., 2002). Walter Reed Army Institute of

Research has successfully reported the production of LAV based vaccine that elicits immune response against all the four serotypes of DENV.

2.8.4Subuint vaccine

Subunit vaccines have also proved to be safer in comparison to live attenuated vaccines but are associated with weak immune stimulation mostly relying on adjuvant or other molecules for boosting immune response adequate for protection (Hem and

HogenEsch, 2007; Ishii and Akira, 2007; Webster and Hill, 2003). In a recent approach an immunogenic portion of DENV Envelope protein (E) was combined with five different adjuvants and effective immune activation was observed with all five formulations (Putnak et al., 2005).

A tetravalent vaccine having bivalent adenovirus constructs (CAdVax-Den12 and

CAdVax-Den34) provided a long-term protection in macaque monkeys by production of neutralizing antibodies against all four serotypes (Raviprakash et al., 2008). In a different study a recombinant tetravalent vaccine having consensus domain III of DENV Envelope protein (E) in combination with aluminum phosphate elicited higher antibody response along with memory immunity in Blab/c mice (Leng et al., 2009).

51

Chapter 2 Review of Literature

2.9 Epitope variation a challenge for DENV vaccine design

DENV is a highly variable virus this variance has resulted in failure of multiple attempts for development of a tetravalent DENV vaccine (Rothman, 2004). Variability varies at the serotype level with high inter-serotype and low intra-serotype variance(Livingston et al., 1995). This serotype specific variance is the main reason for multiple peptide ligands spanning the whole DENV poly-protein with differences of one or several amino acids(Sloan-Lancaster and Allen, 1996), diversity of host immune responses is attributed to these epitope variations (Mongkolsapaya et al., 2006; Welsh and Rothman, 2003).

Advances in immunology have greatly increased the understanding of antigen variation and host immune response associated with different peptide ligands. Antigen variation can result in suppressed, enhanced or neutral host immune response which further increases the need for analysis of antigenic variance for reaching a potent vaccine candidate (Takahashi et al., 1989). For resolving the concerns regarding antigenic variance of DENV a bioinformatics approach can be implemented for ruling out cross reactive and low immunogenic epitopes. A strategy involving T-cell based epitopic prediction will enhance the efficacy of future vaccine tremendously.

2.9.1 DENV T Cell antigenic diversity analysis

Host T cells are targets for mapping and analysis Helper and cytotoxic T lymphocytes mediate cellular immune responses via the T-cell receptors (TCR) that recognize T-cell epitopes presented on cell surfaces by HLA molecules. HLA class I molecules, expressed on the surface of most nucleated cells, present endogenous

52

Chapter 2 Review of Literature

epitopes, synthesized and processed in the 26 cytoplasm, to CD8+ cytotoxic T

Lymphocytes (CTLs) that eventually kill the infected cells(Bjorkman and Parham, 1990;

Shastri et al., 2002).

CD8+ cells are important in conferring immune response against intracellular viruses. On the other hand, HLA class II molecules display exogenously derived epitopes on the surface of professional antigen presenting cells (APCs), such as dendritic cells, B- cells and macrophages, for immune recognition by CD4+ helper T cells. Activated helper

T cells produce secondary signals for activation of both T cells and B cells (Esser et al.,

2003; Pulendran and Ahmed, 2006;Zinkernagel, 2004)

2.9.2 T cell epitope mapping of DENV

Immune studies have demonstrated that both structural and non-structural proteins are able induce T cell mediated immune response (Bashyam et al., 2006; Simmons et al.,

2005). Predominantly helper T-cell epitopic regions are present on the structural proteins where as cytotoxic T-cell epitopes are located on NS proteins (Roehrig, 2003). Studies have shown that human leukocyte antigen (HLA) alleles are the main sites for binding of

DENV T cell epitopes. Association of DENV T cells epitopes with 12 HLA alleles has been established, most of theses epitopes are located on NS3 protein(Screaton and

Mongkolsapaya, 2006; Welsh and Rothman, 2003).

2.9.3 Insilico analysis of DENV T cell epitopes

Recently several studies have shown bioinformatics based DENV T cell epitopes mapping which was later proved by wet lab experiments, therefore bioinformatics is seen as a reliable tool for T cell mapping (Lin et al., 2008b; Wang et

53

Chapter 2 Review of Literature

al., 2008). Advancements in the field of bioinformatics can be utilized by researchers working on host immune interactions and use this knowledge for development of anti- dengue vaccines and therapeutics.

2.9.4 B cell epitope mapping antigenic diversity analysis

Research conducted on DF patients revealed that infection resulted in activation of B cell response after approximately 4 days post infection (Polyreactive, 2011). DENV structural proteins along with NS1 are responsible for host antibody response therefore, identification of B cell epitopes is vital for designing effective vaccine and drugs against

DENV. Characterization of B cell epitopes will also pave the way for development of rapid diagnostic kits and reagents for detection of DENV (Beltramello et al., 2010; Wu et al., 2003). Some of the B cell epitopes were indentified for highly immunogenic DENV proteins (E, prM, C, NS1 and NS4a) however, with high rate of mutations and the presence of several strains makes it an uphill task to fully identify B cell epitopes (Brien et al., 2010; Lin et al., 2012; Steidel et al., 2012).

2.9.5Insilico analysis of DENV B cell epitopes

Previous approaches directed at B cell epitope prediction were focusing at E, NS1 and NS5 proteins (Chiou et al., 2012; Sun et al., 2012). Later studies revealed the importance of C and prM proteins in production of antibody responses, there is still a long way to go for complete B cell epitope mapping of DENV (Dejnirattisai et al.,

2010a). Initially B cell epitopes were identified via techniques for antisera analysis like overlapping synthetic peptides (PEPSCAN) (Aaskov et al., 1989; Innis et al., 1989).

54

Chapter 2 Review of Literature

Several antigenic regions were recognized by recombinant technology as well as protein cleavage by enzymes (Megret et al., 1992; Roehrig et al., 1998).

B cell epitopes can also be identified by displaying fusion peptide on the surface of a phage know as Phage display. This technique has been widely used because it allows conformational changes in the antigenic protein as seen in the virus itself (D'Mello et al.,

1997; Fu et al., 1997;Young et al., 1997). There is a lack of serotype based epitope mapping which is a major factor for achieving a uniform immune response against each serotype in a tetravalent vaccine. Furthermore, emergence of new strains makes B cell epitope mapping a vital component of a future tetravalent DENV vaccine.

Although experimental techniques were successful in defining some B cell epitopes located on DENV proteins however, there are a number of limitations of wet laboratory experiments. Computer based algorithms were able to overcome these limitations and these softwares were able to map B cell epitopes in a very short time with minimum effort(Yang and Yu, 2009; Yasser and Honavar, 2010).

In comparison with T cell epitope prediction, B cell epitope prediction is much intricate owing to a high variability of epitope length as recently described by Kringelum et al reporting that average length of epitopes was 15 amino acid residues (Kringelum et al., 2013). This study explained the fact that exact length of conformational B cell epitope is still not clear however, overall average was 15 residues. The software based epitope mapping has certain limitations like having limited number of epitopes and secondly inability to detect of random peptides as B cell epitopes(Chen et al., 2007a;

EL‐Manzalawy et al., 2008; Sweredoski and Baldi, 2009).

55

Chapter 2 Review of Literature

DF is one of the major global health risk across the globe therefore, anti-dengue vaccine and drug development is vital for safety of people living all over the world.

DENV vaccine development faces major difficulties like antibody-dependent enhancement (ADE) and multiple serotypes, these factors led to the failure of several previous vaccinal strategies (Sabchareon et al., 2012). It will be worthwhile to use an alternative approach utilizing B cell epitope mapping minimizing weak links like cross- reacting antibody responses and variable immune response (Crill et al., 2012;

Dejnirattisai et al., 2010a).

56

Chapter 3 Materials and Methods

Chapter 3

MATERIAL AND METHODS

57

Chapter 3 Materials and Methods

58

Chapter 3 Materials and Methods

59

Chapter 3 Materials and Methods

60

Chapter 3 Materials and Methods

61

Chapter 3 Materials and Methods

62

Chapter 3 Materials and Methods

63

Chapter 3 Materials and Methods

64

Chapter 3 Materials and Methods

65

Chapter 3 Materials and Methods

66

Chapter 3 Materials and Methods

67

Chapter 3 Materials and Methods

68

Chapter 3 Materials and Methods

69

Chapter 4 Results

70

Chapter 4 Results

71

Chapter 4 Results

72

Chapter 4 Results

73

Chapter 4 Results

74

Chapter 4 Results

75

Chapter 4 Results

76

Chapter 4 Results

77

Chapter 4 Results

78

Chapter 4 Results

79

Chapter 4 Results

80

Chapter 4 Results

81

Chapter 4 Results

82

Chapter 4 Results

83

Chapter 4 Results

84

Chapter 4 Results

85

Chapter 4 Results

86

Chapter 4 Results

87

Chapter 4 Results

88

Chapter 4 Results

89

Chapter 4 Results

90

Chapter 4 Results

91

Chapter 4 Results

92

Chapter 4 Results

93

Chapter 4 Results

94

Chapter 4 Results

95

Chapter 4 Results

96

Chapter 4 Results

97

Chapter 4 Results

98

Chapter 4 Results

99

Chapter 4 Results

100

Chapter 4 Results

101

Chapter 4 Results

102

Chapter 4 Results

103

Chapter 4 Results

104

Chapter 4 Results

105

Chapter 4 Results

106

Chapter 4 Results

107

Chapter 4 Results

108

Chapter 4 Results

109

Chapter 4 Results

110

Chapter 4 Results

111

Chapter 4 Results

112

Chapter 4 Results

113

Chapter 4 Results

114

Chapter 4 Results

115

Chapter 4 Results

116

Chapter 4 Results

117

Chapter 4 Results

118

Chapter 4 Results

119

Chapter 4 Results

120

Chapter 4 Results

121

Chapter 4 Results

a

b

122

Chapter 4 Results a b

123

Chapter 4 Results

a b

124

Chapter 4 Results

125

Chapter 4 Results

126

Chapter 4 Results

127

Chapter 4 Results

128

Chapter 4 Results

129

Chapter 4 Results

130

Chapter 4 Results

131

Chapter 4 Results

132

Chapter 4 Results

133

Chapter 4 Results

134

Chapter 4 Results

135

Chapter 4 Results

136

Chapter 4 Results

137

Chapter 4 Results

138

Chapter 4 Results

139

Chapter 4 Results

140

Chapter 4 Results

141

Chapter 4 Results

142

Chapter 4 Results

143

Chapter 4 Results

144

Chapter 4 Results

145

Chapter 4 Results

146

Chapter 4 Results

147

Chapter 4 Results

148

Chapter 4 Results

149

Chapter 4 Results

150

Chapter 4 Results

151

Chapter 4 Results

152

Chapter 4 Results

153

Chapter 4 Results

154

Chapter 4 Results

155

Chapter 4 Results

156

Chapter 4 Results

157

Chapter 4 Results

158

Chapter 4 Results

159

Chapter 4 Results

160

Chapter 4 Results

161

Chapter 4 Results

162

Chapter 4 Results

163

Chapter 4 Results

164

Chapter 4 Results

165

Chapter 4 Results

166

Chapter 4 Results

167

Chapter 4 Results

168

Chapter 4 Results

169

Chapter 4 Results

170

Chapter 4 Results

171

Chapter 4 Results

172

Chapter 5 Discussion

Chapter 5

DISCUSSION

Amongst arthropods borne viral infections the leading cause is Dengue virus.

According to recent estimates annual cases range from 100 to 200 million and with approximately 2 billion people living in high risk zones (Murray et al., 2013). Children living in these endemic regions are at high risk owing to disease severity and high mortality associated with DHF/DSS (Poovorawan et al., 2006). These factors further increase need for development of a vaccine and inhibitors for protecting general population and reducing annual mortality amongst children and adults.Molecular research conducted in viral replication and protein synthesis revealed several potential targets for anti-viral therapeutic development.

According to some recent studies, inhibition of target enzymes and proteins stemmed viral growth. siRNA also provides a very safe alternative to chemotherapeutic agents because of its limited side effects and its specificity for viral genomes. However limitations like lack of effective delivery mechanisms, time dependent reduction in RNA interference and expensiveness are major hurdles in the use of siRNA as possible anti- dengue therapeutic agent.

Last twenty years have seen DENV becoming a major global threat therefore; drastic measures are required for limiting this viral pathogen. Scientists have been working on different vaccine and therapeutic approaches for against DENV since the emergence of the disease. However advances in molecular biology helped in understanding various aspects of DENV replication and host interactions, this

173

Chapter 5 Discussion

information led to the development of new inhibitors and vaccines. The propagation of

DENV in mosquitoes as well as humans was explained by a number of biologist which suggested that blood meal of an infected human individual results in replication of DENV in mosquitoes gut region(Hidari and Suzuki, 2011). After achieving high titers DENV travels to the salivary gland and it is transmitted to a healthy person via a new blood meal. Inside the human host DENV is taken up by antigen presenting cells (APCs) via endocytosis involving cellular protein clathrin(Sun and Kochel, 2013). DENV entry was explained in depth by Alhoot et al 2011 and suggested that clathrin mediated endocytosis was crucial for DENV internalization in the host cell (Alhoot et al., 2011). Receptor studies revealed that CD14 receptor expressed on the membranes of immune cells play central role in binding and endocytosis of DENV (Alhoot et al., 2011). In one of the inhibitory approach CD14 expression was suppressed by siRNAs which led to massive reduction in the expression of cellular CD14 receptors. When immune cells with limited expression of CD14 were presented with DENV challenge it was observed that internalization of DENV was drastically reduced (Alhoot et al., 2011). Although RNAi is a very effective tool for gene silencing however there are some limitations in RNAi technology, one of these limitation is host cell responses for degrading dsRNA. To tackle this concern a study was conducted which was able to enhance the knock down efficiency of siRNA and provided protection against host cell responses (Fischer and James, 2004).

In mosquitoes DENV can effectively replicate in several locations this is one of the reason which lead to the development of C6/36 cell line (Sakoonwatanyoo et al.,

2006). These C6/36 cells are mosquito cells which allow replication of DENV and yield high titer of DENV in vitrostudies. Xinweiet al in 2010 conducted a study for inhibiting

174

Chapter 5 Discussion

DENV replication in C6/36 cells, when C6/36 cells treated with siRNAwere challenged with DENV a huge drop in DENV RNA was observed in addition to reduction in viral titer cell survival rate increased tremendously (Wu et al., 2010).

With the recent advancements in the field of molecular biology,siRNA technology has become a leading tool in gene therapy with applications like up regulation and down regulation of certain target genes. Based on this enormous potential siRNA technology is being extensively used for anti-virus drug development. With advantages like specificity, minimum or no toxicity and efficient mode of action siRNAs are one of the important components of biopharmaceuticals. Recent studies suggest that siRNAs can be used for blocking virus transmission (Joost Haasnoot et al., 2003). DENV carrying a single stranded RNA as the genome in the cytoplasm is a probable target for different

RNAi based inhibition mechanisms.

Arboviruses with RNA genome require an intermediate dsRNA molecule which can trigger host cell RNAi pathway (Blair and Olson, 2015). Cellular enzyme DICER chopsdsRNA and generates several small interfering RNAs, these 20-24 base pair siRNAs interact with argonate containing RISC (RNA induced silencing complex). This complex causes the unwinding of dsRNA and one of the strand of this dsRNA serves as a target locator, with the help of this sequence RISC binds with the target mRNA. In the next step the endonuclease activity takes place and cuts the target mRNA resulting in suppression of that specific gene (Dunoyer et al., 2004). As RNAi has been a part of innate immunity of most eukaryotic cells therefore this aspect was utilized by biologist for development of anti-viral inhibitors. This discovery led to the application of RNAi in several experiments involving gene silencing and pathogen inhibition. In recent times

175

Chapter 5 Discussion

several human viruses were successfully targeted by RNAi which proves that RNAi is an effective tool in modern therapeutics (Bogerd et al., 2014).

In the current study we observed that synthetic siRNAswhich targeting the highly conserved regions of DENV3 genome can effectively inhibit DENV3 in Vero-81 cell lines. These conserved regions of DENV genome are potential targets for effective designing and targeting, moreover this information may be useful for future studies involving genome based inhibition of DENV. Three regions (3' UTR, 5' UTR and structural genes) were specifically targeted in this study since these regions were found to be highly conserved and also play a key role in DENV replication and assembly.3'UTR is approximately 450 nucleotide in length without a poly A tail with several conserve structures. These folds at 3' UTR results in the formation of conserve looped structures known as stem loop (3'SL), in depth studies conducted on the properties of this 3' SL in different arbovirusesrevealed that 3'SL is a key component of DENV replication (Alvarez et al., 2005a). In comparison to SL other components of 3'UTR are still not explained in detail and the exact function of other conserve regions are still a mystery (Wei et al.,

2009). In addition to SL regions various softwares have been able to predict two further folded structures A2 and A3. When these two folds were thoroughly studied via available algorithms, it was observed that both folds had conserved regions CS2 and RCS2

(Manzano et al., 2011). 3'UTR is significantly important in DENV replication this was proved by experiments in which deletions in the 3'UTR greatly hindered DENV replication and also resulted in defective virion formation (Tilgner et al., 2005). The exact role of 3'UTR in DENV replication is still not fully known, inhibitors could be designed only if the whole mechanism is revealed. Thus, 3'UTR has critical role in

176

Chapter 5 Discussion

DENV replication and disruption of conserve domains in the 3'UTR will result in impaired DENV replication and dysfunctional virus production.

5'UTR is highly conserved in all the serotypes, it has contains approximately 100 base pairs. In depth analysis revealed that 5'UTR has a essentialrole in DENV replication

(Filomatori et al., 2006). Studies have shown that 5'UTR elements form a stem loop secondary structures and these structures are conserved amongst several members of arboviruses(Gebhard et al., 2011). Deletions in the 5'UTR region resulted in defective protein production and disruption of normal cell replication. It was seen that 5'UTR elements were responsible for binding of RNA with host proteins that are responsible for translation of protein (Sirigulpanit et al., 2007). Thus 5'UTR is pivotal for DENV replication but experiments have also suggested that 5'UTR is also involved in viral assembly (Roby et al., 2014).

All six siRNAs targeting DENV3 genome underwent strict scrutiny and only those siRNAs were selected which were able to fulfill all vital parameters for siRNA designing. Regions targeted included 3'UTR, 5'UTR and structural genes (C, prM and E) in DENV3 genome, all three regions are vital for maintaining replication cycle of

DENV3. siRNA designed from 3'UTR showed a significant reduction in the cellular

DENV3 level, which was confirmed by IFA and real time qPCR. A previous study conducted by Diego et al also demonstrated that deletions in 3'UTR resulted in several folds decrease in DENV titer. Results showed DENV3UTR3'siRNA2 was highly effective in limiting DENV3 replication when compared with other five siRNAs which were similarly tested for inhibition study.

177

Chapter 5 Discussion

Foci counts revealed that DENV3UTR5'siRNA designed from 5UTR was also able to reduce DENV3 up to 65% 48 hours post DENV3 challenge. Synthetic siRNAs(DENV3SsiRNA1 and DENV3SsiRNA2)targeting the structural region had a limited triumph in limiting viral replication. As previously discussed UTRs are key elements in viral replication and genome organization, this can be one of the explanation for higher inhibition efficacy of siRNAs designed to target UTRs in DENV genome.

RNA disruption at UTRs via RNAi can significantly hinder viral replication and this can lead to reduce and faulty genome replication as well as protein synthesis.

Experiments conducted on DENV3 in Vero-81 cells demonstrated that synthetic siRNAs can effectively inhibit DENV3 and proved to be an efficient molecular tool for development of anti-dengue inhibitor. Although there are concerns regarding short half life of siRNAs and targeted delivery, however scientists are developing new technologies to overcome these issues. Yuen et al were able to achieve targeted delivery via combining nanotechnology and liposomes, this approach was successful in vivo models. Futures studies involving siRNAs designed in this study will elucidate the role of molecular medicine against DENV.

Previously most RNAi approaches were based on random targeting of DENV genome as well as host factors associated with entry and replication of DENV. This approach was useful for inhibiting DENV however there was still need for identifying highly conserved regions that can be efficiently targeted for development of lasting inhibitors. siRNAs were designed from conserved regions located in DENV2 genome for inhibition studies. These conserved regions were selected for persistent effectiveness of

RNAi, as other regions could undergo mutations which can render the siRNAsineffective.

178

Chapter 5 Discussion

This information regarding DENV2conserved regions would help future endeavors in the anti-dengue drug development and vaccine production.

Most of the previous research concerning DENV therapeutics and vaccine formulation was conducted by utilizing DENVs propagated in C6/36 cells. That is why

C6/36 cells are most abundantly used cell line for DENV propagation, however progeny

DENVs are a diverse population of immature and mature DENV due to inefficient Furin related conversion of prMto M (Richter et al., 2014; Zybert et al., 2008). This mixed population is later on used for anti-dengue therapeutic testing as well as neutralization studies involving vaccine candidates.

Dendritic cells (DCs) are important component of our immune system and provide immunity against several pathogens however these DCs serve as target sites for

DENV replication (Marovich et al., 2001). DENV can effectively replicate in DCs, consequently one of the study was specifically aimed for targeting DENVs in infected

DCs. A complex of peptide and siRNA was introduced in infected DCs for targeting

DENV E gene; this peptide and nucleic acid fusion significantly decrease DENV replication. This fusion complex was also tested in mice models grafted with human cells results showed that this complex was able to knock down DENV and DENV challenged mice survived (Subramanya et al., 2010).Although studies have shown that immature and mature DENVs are infectious and both respond similarly to anti viral therapeutics(Zybert et al., 2008). However, in human body replication of DENV mostly occurs in DCs yielding mostly mature virions. Therefore, to maximize the efficacy of siRNA for future in vivo studies experiments were done on mature DENVs. Mature DENV2s were propagated in U937 DC-SIGN cells; these cells had adequate Furin to yield higher

179

Chapter 5 Discussion

proportion of mature DENV2. To confirm that the population of DENV2s derived from

DC SIGNS was mostly mature western blot was performed for detection of prMprotein, samples showing insignificant levels of prM were selected for further experimentations.

Mature DENV2 were targeted by six siRNAs designed from UTRs (3' and 5'), these regions were highly conserved and had central role in DENV replication and assembly. Results revealed that DENV2SsiRNA2 was able to reduce DENV2 significantly, 85-90% reduction in DENV2titer was observed in cells transfected by

DENV2SsiRNA2. Promising results were also shown by DENV2UTR3'siRNA2 which was able to drop DENV2 titer up to 55%. However remaining four siRNAs did not had any significant effect on DENV2 titers, foci counts revealed that these four siRNAs had similar number in comparison with control groups. Thus only two siRNAs

(DENV2SsiRNA2 and DENV2UTR3'siRNA2 were effective in limiting DENV replication. These results show that mature DENV2 can be effectively targeted by RNAi, with appropriate delivery mechanism and sustained release RNAi can be an effective anti-viral therapeutic agent in future.

Currently several studies are being conducted for development of therapeutic agents utilizing RNAi technology but several factors can lead to reduced efficiency of these siRNA derived approaches. Some of these include short half life and a swift degradation in the cells. This rapid unavailability can lead to recurrence as well as ineffectiveness in complete knock out of pathogen. One of the major concern regarding siRNAtherapy is targeted delivery because it is vital for siRNA to reach target cells to knock out gene of interest. For targeted delivery researchers are focusing on liposome

180

Chapter 5 Discussion

and nanotechnology these approaches have yielded promising results and several of these highly specific siRNAs have made it to clinical trials.

Cross-protective tetravalent vaccine with a constant immune can be a major breakthrough against DF/DHF, since a protective vaccine employed before the onset of rainy season will ensure safety of the general population. Numerous attempts for reaching a DENVvaccine have resulted in failure because of multiple factors including cross- reactivity amongst serotypes (Mahalingam et al., 2013). Latest approaches in vaccine development like DNA vaccines and sub-unit vaccine proved to be useful and many of these vaccinal candidates are in clinical trial phases (Kanesa-Thasan et al., 2003).

Tetravalent vaccines can also provide protection against all four serotypes. These approaches exploiting both virus and host targets can ultimately lead to the development of DENV inhibitors and vaccines as treatment and prophylactic options.

Dengue virus has three structural genes which are essential for viral stability and interaction with host cell via receptor mediated endocytosis. In humans majority of antibodies are produced against E protein, as E is highly immunogenic that is why it is one of the major component of vaccine strategies (Lin et al., 2012).DENV structural proteins play a key role in binding with host cell receptors as well as maintaining virus integrity. Although all three proteins are immunogenic however, majority of antibodies are produced against E protein. Antibodies are also generated against C and prM but studies have revealed that several prM antibodies are cross-reactive weakly neutralizing.

Two types are of antibodies are produced against DENV structural proteins strongly neutralizing antibodies predominantly targeting the structural region and cross reactive weakly neutralizing antibodies generated against the prM/M regions

181

Chapter 5 Discussion

(Dejnirattisai et al., 2010b). Thus, structural proteins play a central role in maintaining the viral integrity and targeting structural genes will result in dysfunctional virion production lacking stability and infectivity. Therefore, a vaccine candidate should have only those eptitopes which can generate strongly neutralizing antibodies for providing maximum protection against DENV. A perfect vaccine should provide cross protection against all serotypes from every corner of globe. With the discovery of fifth DENV serotype and several regional variants the importance of phylogenetics has boosted tremendously, there is an urgent need for evaluating these changes for reaching a global

DENV vaccine.

For evaluating the linkages of Pakistani DENV2 structural genes with worldwide sequences insilico study was performed. Phylogenetic trees revealed that Pakistani

DENV2 is similar to Indian DENV2 strain which points to the fact that geographical proximity can result in the spread of infections in related populations. In 2010 outbreak, thousands of cases were reported in the city of Lahore which is located near the Indian border and several hundred people travel across both countries via Lahore, which could have led to the simultaneously outbreak in both countries. Moreover, this data will help future researchers working on the development of universal DENV vaccine to ensure that vaccine is equally effective for both countries of this region.

Antigenic sites located on structural proteins induce the host immune response, these include both B and T cell epitopes. However, most vaccines rely on humoral immune response for neutralizing of a specific pathogen for this reason B cell epitopes of

Pakistani DENV2 isolates were predicted in the current study. Generally there are two main reasons for B cell epitope prediction, for development of vaccine or a diagnostic kit.

182

Chapter 5 Discussion

Viral infections can be diagnosed by using antibodies targeting antigenic domains of a pathogen. Small peptide based vaccines can be used for prophylaxis and curative purposes, as these peptides are highly antigenic in nature. By applying these technique problems like production of cross-reactive weakly neutralizing antibodies can be addressed without the possibility of enhancing infection. In the present study, insilico analysis comprising of features like inter-molecular contact, antigenicity, residue solvent accessibility, flexibility, surface accessibility and spatial distribution were performed on all three structural DENV2 proteins. By using these linear epitopes monoclonal neutralizing antibodies can be generated and thus result in the production of an effective

DENV vaccine.

183

Chapter 6 References

Chapter 6

REFERENCES

Aaskov, J., Geysen, H. and Mason, T. (1989). Serologically defined linear epitopes in the envelope protein of dengue 2 (Jamaica strain 1409). Archives of virology, 105 (3-4): 209- 221. Adams, S., Broom, A., Sammels, L., Hartnett, A., Howard, M., Coelen, R., Mackenzie, J. and Hall, R. (1995). Glycosylation and antigenic variation among Kunjin virus isolates. Virology, 206 (1): 49-56. Adelman, Z., Blair, C., Carlson, J., Beaty, B. and Olson, K. (2001). Sindbis virus‐induced silencing of dengue viruses in mosquitoes. Insect molecular biology, 10 (3): 265-273. Akram, D. S., Igarashi, A. and Takasu, T. (1998). Dengue virus infection among children with undifferentiated fever in Karachi. The Indian Journal of Pediatrics, 65 (5): 735-740. Alcaraz-Estrada, S. L., Yocupicio-Monroy, M. and Del Angel, R. M. (2010). Insights into dengue virus genome replication. Future Virology, 5 (5): 575-592. Alen, M. M., Kaptein, S. J., De Burghgraeve, T., Balzarini, J., Neyts, J. and Schols, D. (2009). Antiviral activity of carbohydrate-binding agents and the role of DC-SIGN in dengue virus infection. Virology, 387 (1): 67-75. Aleshin, A. E., Shiryaev, S. A., Strongin, A. Y. and Liddington, R. C. (2007). Structural evidence for regulation and specificity of flaviviral proteases and evolution of the Flaviviridae fold. Protein science, 16 (5): 795-806. Alhoot, M. A., Wang, S. M. and Sekaran, S. D. (2011). Inhibition of dengue virus entry and multiplication into monocytes using RNA interference. PLoS Negl Trop Dis, 5 (11): e1410- e1410. Allison, S. L., Schalich, J., Stiasny, K., Mandl, C. W. and Heinz, F. X. (2001). Mutational evidence for an internal fusion peptide in flavivirus envelope protein E. Journal of virology, 75 (9): 4268-4275. Almeida, R. and Allshire, R. C. (2005). RNA silencing and genome regulation. Trends in cell biology, 15 (5): 251-258. Alvarez, D. E., Ezcurra, A. L. D. L., Fucito, S. and Gamarnik, A. V. (2005a). Role of RNA structures present at the 3′ UTR of dengue virus on translation, RNA synthesis, and viral replication. Virology, 339 (2): 200-212. Alvarez, D. E., Filomatori, C. V. and Gamarnik, A. V. (2008). Functional analysis of dengue virus cyclization sequences located at the 5′ and 3′ UTRs. Virology, 375 (1): 223-235. Alvarez, D. E., Lodeiro, M. F., Ludueña, S. J., Pietrasanta, L. I. and Gamarnik, A. V. (2005b). Long- range RNA-RNA interactions circularize the dengue virus genome. Journal of virology, 79 (11): 6631-6643. Amberg, S. M. and Rice, C. M. (1999). Mutagenesis of the NS2B-NS3-mediated cleavage site in the flavivirus capsid protein demonstrates a requirement for coordinated processing. Journal of virology, 73 (10): 8083-8094. An, J., Zhou, D.-S., Zhang, J.-L., Morida, H., Wang, J.-L. and Yasui, K. (2004). Dengue-specific CD8+ T cells have both protective and pathogenic roles in dengue virus infection. Immunology letters, 95 (2): 167-174.

184

Chapter 6 References

Anders, K. L., Nguyet, N. M., Chau, N. V. V., Hung, N. T., Thuy, T. T., Farrar, J., Wills, B., Hien, T. T. and Simmons, C. P. (2011). Epidemiological factors associated with dengue shock syndrome and mortality in hospitalized dengue patients in Ho Chi Minh City, Vietnam. The American journal of tropical medicine and hygiene, 84 (1): 127-134. Andrews, B., Theofilopoulos, A., Peters, C., Loskutoff, D., Brandt, W. and Dixon, F. (1978). Replication of dengue and junin viruses in cultured rabbit and human endothelial cells. Infection and immunity, 20 (3): 776-781. Araújo, J. M., Nogueira, R. M., Schatzmayr, H. G., Paolo, M. D. A. and Bello, G. (2009). Phylogeography and evolutionary history of dengue virus type 3. Infection, Genetics and Evolution, 9 (4): 716-725. Avirutnan, P., Punyadee, N., Noisakran, S., Komoltri, C., Thiemmeca, S., Auethavornanan, K., Jairungsri, A., Kanlaya, R., Tangthawornchaikul, N. and Puttikhunt, C. (2006). Vascular leakage in severe dengue virus infections: a potential role for the nonstructural viral protein NS1 and complement. Journal of Infectious Diseases, 193 (8): 1078-1088. Bagga, S., Bracht, J., Hunter, S., Massirer, K., Holtz, J., Eachus, R. and Pasquinelli, A. E. (2005). Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell, 122 (4): 553-563. Bai, F., Town, T., Pradhan, D., Cox, J., Ledizet, M., Anderson, J. F., Flavell, R. A., Krueger, J. K., Koski, R. A. and Fikrig, E. (2007). Antiviral peptides targeting the west nile virus envelope protein. Journal of virology, 81 (4): 2047-2055. Balsitis, S. J., Williams, K. L., Lachica, R., Flores, D., Kyle, J. L., Mehlhop, E., Johnson, S., Diamond, M. S., Beatty, P. R. and Harris, E. (2010). Lethal antibody enhancement of dengue disease in mice is prevented by Fc modification. PLoS Pathog, 6 (2): e1000790. Barbas, C. F., Heine, A., Zhong, G., Hoffmann, T., Gramatikova, S., Björnestedt, R., List, B., Anderson, J., Stura, E. A. and Wilson, I. A. (1997). Immune versus natural selection: antibody aldolases with enzymic rates but broader scope. Science, 278 (5346): 2085- 2092. Barrera, R., Delgado, N., Jiménez, M. and Valero, S. (2002). Eco-epidemiological factors associated with hyperendemic dengue haemorrhagic fever in Maracay City, Venezuela. Dengue Bulletin, 26: 84-95. Bartelma, G. and Padmanabhan, R. (2002). Expression, purification, and characterization of the RNA 5′-triphosphatase activity of dengue virus type 2 nonstructural protein 3. Virology, 299 (1): 122-132. Bashyam, H. S., Green, S. and Rothman, A. L. (2006). Dengue virus-reactive CD8+ T cells display quantitative and qualitative differences in their response to variant epitopes of heterologous viral serotypes. The Journal of Immunology, 176 (5): 2817-2824. Bazan, J. F. and Fletterick, R. J. (1989). Detection of a trypsin-like serine protease domain in flaviviruses and pestviruses. Virology, 171 (2): 637-639. Beaumier, C. M. and Rothman, A. L. (2009). Cross-reactive memory CD4+ T cells alter the CD8+ T-cell response to heterologous secondary dengue virus infections in mice in a sequence-specific manner. Viral immunology, 22 (3): 215-219. Beltramello, M., Williams, K. L., Simmons, C. P., Macagno, A., Simonelli, L., Quyen, N. T. H., Sukupolvi-Petty, S., Navarro-Sanchez, E., Young, P. R. and De Silva, A. M. (2010). The human immune response to Dengue virus is dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity. Cell host & microbe, 8 (3): 271-283.

185

Chapter 6 References

Benarroch, D., Egloff, M.-P., Mulard, L., Guerreiro, C., Romette, J.-L. and Canard, B. (2004). A structural basis for the inhibition of the NS5 dengue virus mRNA 2′-O-methyltransferase domain by ribavirin 5′-triphosphate. Journal of Biological Chemistry, 279 (34): 35638- 35643. Bente, D. A. and Rico-Hesse, R. (2006). Models of dengue virus infection. Drug Discovery Today: Disease Models, 3 (1): 97-103. Bjorkman, P. J. and Parham, P. (1990). Structure, function, and diversity of class I major histocompatibility complex molecules. Annual review of biochemistry, 59 (1): 253-288. Blair, C. D. and Olson, K. E. (2015). The Role of RNA Interference (RNAi) in Arbovirus-Vector Interactions. Viruses, 7 (2): 820-843. Bogerd, H. P., Skalsky, R. L., Kennedy, E. M., Furuse, Y., Whisnant, A. W., Flores, O., Schultz, K. L., Putnam, N., Barrows, N. J. and Sherry, B. (2014). Replication of many human viruses is refractory to inhibition by endogenous cellular microRNAs. Journal of virology, 88 (14): 8065-8076. Boonnak, K., Slike, B. M., Burgess, T. H., Mason, R. M., Wu, S.-J., Sun, P., Porter, K., Rudiman, I. F., Yuwono, D. and Puthavathana, P. (2008). Role of dendritic cells in antibody- dependent enhancement of dengue virus infection. Journal of virology, 82 (8): 3939- 3951. Bosch, I., Xhaja, K., Estevez, L., Raines, G., Melichar, H., Warke, R. V., Fournier, M. V., Ennis, F. A. and Rothman, A. L. (2002). Increased production of interleukin-8 in primary human monocytes and in human epithelial and endothelial cell lines after dengue virus challenge. Journal of virology, 76 (11): 5588-5597. Boulton, R. and Westaway, E. (1977). Togavirus RNA: Reversible effect of urea on genomes and absence of subgenomic viral RNA in Kunjin virus-infected cells. Archives of virology, 55 (3): 201-208. Bower, M. J., Cohen, F. E. and Dunbrack, R. L. (1997). Prediction of protein side-chain rotamers from a backbone-dependent rotamer library: a new homology modeling tool. Journal of molecular biology, 267 (5): 1268-1282. Brien, J. D., Austin, S. K., Sukupolvi-Petty, S., O'brien, K. M., Johnson, S., Fremont, D. H. and Diamond, M. S. (2010). Genotype-specific neutralization and protection by antibodies against dengue virus type 3. Journal of virology, 84 (20): 10630-10643. Brinton, M. A., Kurane, I., Mathew, A., Zeng, L., Shi, P. Y., Rothman, A. and Ennis, F. A. (1998). Immune mediated and inherited defences against flaviviruses. Clinical and diagnostic virology, 10 (2): 129-139. Cahour, A., Falgout, B. and Lai, C. (1992). Cleavage of the dengue virus polyprotein at the NS3/NS4A and NS4B/NS5 junctions is mediated by viral protease NS2B-NS3, whereas NS4A/NS4B may be processed by a cellular protease. Journal of virology, 66 (3): 1535- 1542. Caplen, N. J., Zheng, Z., Falgout, B. and Morgan, R. A. (2002). Inhibition of viral gene expression and replication in mosquito cells by dsRNA-triggered RNA interference. Molecular Therapy, 6 (2): 243-251. Carroll, S. S., Ludmerer, S., Handt, L., Koeplinger, K., Zhang, N. R., Graham, D., Davies, M.-E., Maccoss, M., Hazuda, D. and Olsen, D. B. (2009). Robust antiviral efficacy upon administration of a nucleoside analog to virus-infected chimpanzees. Antimicrobial agents and chemotherapy, 53 (3): 926-934. Chambers, T. J., Hahn, C. S., Galler, R. and Rice, C. M. (1990). Flavivirus genome organization, expression, and replication. Annual Reviews in Microbiology, 44 (1): 649-688.

186

Chapter 6 References

Chareonsirisuthigul, T., Kalayanarooj, S. and Ubol, S. (2007). Dengue virus (DENV) antibody- dependent enhancement of infection upregulates the production of anti-inflammatory cytokines, but suppresses anti-DENV free radical and pro-inflammatory cytokine production, in THP-1 cells. Journal of general virology, 88 (2): 365-375. Chau, T. N. B., Hieu, N. T., Anders, K. L., Wolbers, M., Hieu, L. T. M., Hien, T. T., Hung, N. T., Farrar, J., Whitehead, S. and Simmons, C. P. (2009). Dengue virus infections and maternal antibody decay in a prospective birth cohort study of Vietnamese infants. Journal of Infectious Diseases, 200 (12): 1893-1900. Chen, C.-J., Kuo, M.-D., Chien, L.-J., Hsu, S.-L., Wang, Y.-M. and Lin, J.-H. (1997a). RNA-protein interactions: involvement of NS3, NS5, and 3'noncoding regions of Japanese encephalitis virus genomic RNA. Journal of virology, 71 (5): 3466-3473. Chen, J., Liu, H., Yang, J. and Chou, K.-C. (2007a). Prediction of linear B-cell epitopes using amino acid pair antigenicity scale. Amino acids, 33 (3): 423-428. Chen, L., Ewing, D., Subramanian, H., Block, K., Rayner, J., Alterson, K. D., Sedegah, M., Hayes, C., Porter, K. and Raviprakash, K. (2007b). A heterologous DNA prime-Venezuelan equine encephalitis virus replicon particle boost dengue vaccine regimen affords complete protection from virus challenge in cynomolgus macaques. Journal of virology, 81 (21): 11634-11639. Chen, M.-C., Lin, C.-F., Lei, H.-Y., Lin, S.-C., Liu, H.-S., Yeh, T.-M., Anderson, R. and Lin, Y.-S. (2009). Deletion of the C-terminal region of dengue virus nonstructural protein 1 (NS1) abolishes anti-NS1-mediated platelet dysfunction and bleeding tendency. The Journal of Immunology, 183 (3): 1797-1803. Chen, Y.-C., Wang, S.-Y. and King, C.-C. (1999). Bacterial lipopolysaccharide inhibits dengue virus infection of primary human monocytes/macrophages by blockade of virus entry via a CD14-dependent mechanism. Journal of virology, 73 (4): 2650-2657. Chen, Y., Maguire, T., Hileman, R. E., Fromm, J. R., Esko, J. D., Linhardt, R. J. and Marks, R. M. (1997b). Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nature medicine, 3 (8): 866-871. Chiou, S.-S., Fan, Y.-C., Crill, W. D., Chang, R.-Y. and Chang, G.-J. J. (2012). Mutation analysis of the cross-reactive epitopes of Japanese encephalitis virus envelope glycoprotein. Journal of General Virology, 93 (Pt 6): 1185-1192. Chu, J., Rajamanonmani, R., Li, J., Bhuvanakantham, R., Lescar, J. and Ng, M.-L. (2005). Inhibition of West Nile virus entry by using a recombinant domain III from the envelope glycoprotein. Journal of General Virology, 86 (2): 405-412. Chu, J. and Yang, P. L. (2007). c-Src protein kinase inhibitors block assembly and maturation of dengue virus. Proceedings of the National Academy of Sciences, 104 (9): 3520-3525. Churdboonchart, V., Bhamarapravati, N. and Futrakul, P. (1983). Crossed immunoelectrophoresis for the detection of split products of the third complement in dengue hemorrhagic fever. I. Observations in patients' plasma. The American journal of tropical medicine and hygiene, 32 (3): 569-576. Cleaves, G. R. and Dubin, D. T. (1979). Methylation status of intracellular dengue type 2 40 S RNA. Virology, 96 (1): 159-165. Cologna, R., Armstrong, P. M. and Rico-Hesse, R. (2005). Selection for virulent dengue viruses occurs in humans and mosquitoes. Journal of virology, 79 (2): 853-859. Crill, W. D., Hughes, H. R., Trainor, N. B., Davis, B. S., Whitney, M. T. and Chang, G.-J. J. (2012). Sculpting humoral immunity through dengue vaccination to enhance protective immunity. Frontiers in immunology, 3.

187

Chapter 6 References

Crill, W. D. and Roehrig, J. T. (2001). Monoclonal antibodies that bind to domain III of dengue virus E glycoprotein are the most efficient blockers of virus adsorption to Vero cells. Journal of virology, 75 (16): 7769-7773. Cui, T., Sugrue, R. J., Xu, Q., Lee, A. K., Chan, Y.-C. and Fu, J. (1998). Recombinant dengue virus type 1 NS3 protein exhibits specific viral RNA binding and NTPase activity regulated by the NS5 protein. Virology, 246 (2): 409-417. D'mello, F., Partidos, C. D., Steward, M. W. and Howard, C. R. (1997). Definition of the primary structure of hepatitis B virus (HBV) pre-S hepatocyte binding domain using random peptide libraries. Virology, 237 (2): 319-326. Da Glória Teixeira, M., Da Conceicao Nascimento Costa, M., Guerra, Z. and Barreto, M. L. (2002). Dengue in Brazil: situation-2001 and trends. Dengue Bulletin, 26: 70-76. De Nova-Ocampo, M., Villegas-Sepúlveda, N. and Del Angel, R. M. (2002). Translation elongation factor-1α, La, and PTB interact with the 3′ untranslated region of dengue 4 virus RNA. Virology, 295 (2): 337-347. prM De Paula, S. O., Lima, D. M., De Oliveira França, R. F., Gomes-Ruiz, A. C. and Da Fonseca, B. a. L. (2008). A DNA vaccine candidate expressing dengue-3 virus and E proteins elicits neutralizing antibodies and protects mice against lethal challenge. Archives of virology, 153 (12): 2215-2223. Deas, T. S., Binduga-Gajewska, I., Tilgner, M., Ren, P., Stein, D. A., Moulton, H. M., Iversen, P. L., Kauffman, E. B., Kramer, L. D. and Shi, P.-Y. (2005). Inhibition of flavivirus infections by antisense oligomers specifically suppressing viral translation and RNA replication. Journal of virology, 79 (8): 4599-4609. Dejnirattisai, W., Jumnainsong, A., Onsirisakul, N., Fitton, P., Vasanawathana, S., Limpitikul, W., Puttikhunt, C., Edwards, C., Duangchinda, T. and Supasa, S. (2010a). Cross-reacting antibodies enhance dengue virus infection in humans. Science, 328 (5979): 745-748. Dejnirattisai, W., Jumnainsong, A., Onsirisakul, N., Fitton, P., Vasanawathana, S., Limpitikul, W., Puttikhunt, C., Edwards, C., Duangchinda, T. and Supasa, S. (2010b). Enhancing cross- reactive anti-prM dominates the human antibody response in dengue infection. Science (New York, NY), 328 (5979). Dong, H., Ray, D., Ren, S., Zhang, B., Puig-Basagoiti, F., Takagi, Y., Ho, C. K., Li, H. and Shi, P.-Y. (2007). Distinct RNA elements confer specificity to flavivirus RNA cap methylation events. Journal of virology, 81 (9): 4412-4421. Dunham, E. J. and Holmes, E. C. (2007). Inferring the timescale of dengue virus evolution under realistic models of DNA substitution. Journal of molecular evolution, 64 (6): 656-661. Dunoyer, P., Lecellier, C.-H., Parizotto, E. A., Himber, C. and Voinnet, O. (2004). Probing the microRNA and small interfering RNA pathways with virus-encoded suppressors of RNA silencing. The Plant Cell, 16 (5): 1235-1250. Edelman, R. (2007). Dengue vaccines approach the finish line. Clinical Infectious Diseases, 45 (Supplement 1): S56-S60. Edelman, R., Wasserman, S. S., Bodison, S. A., Putnak, R. J., Eckels, K. H., Tang, D., Kanesa- Thasan, N., Vaughn, D. W., Innis, B. L. and Sun, W. (2003). Phase I trial of 16 formulations of a tetravalent live-attenuated dengue vaccine. The American journal of tropical medicine and hygiene, 69 (6 suppl): 48-60. Egloff, M. P., Benarroch, D., Selisko, B., Romette, J. L. and Canard, B. (2002). An RNA cap (nucleoside‐2′‐O‐)‐methyltransferase in the flavivirus RNA polymerase NS5: crystal structure and functional characterization. The EMBO journal, 21 (11): 2757-2768.

188

Chapter 6 References

Ekonomiuk, D., Su, X.-C., Ozawa, K., Bodenreider, C., Lim, S. P., Yin, Z., Keller, T. H., Beer, D., Patel, V. and Otting, G. (2009). Discovery of a non-peptidic inhibitor of West Nile virus NS3 protease by high-throughput docking. PLoS Negl Trop Dis, 3 (1): e356. El‐Manzalawy, Y., Dobbs, D. and Honavar, V. (2008). Predicting linear B‐cell epitopes using string kernels. Journal of molecular recognition, 21 (4): 243-255. Elbashir, S. M., Lendeckel, W. and Tuschl, T. (2001). RNA interference is mediated by 21-and 22- nucleotide RNAs. Genes & development, 15 (2): 188-200. Erbel, P., Schiering, N., D'arcy, A., Renatus, M., Kroemer, M., Lim, S. P., Yin, Z., Keller, T. H., Vasudevan, S. G. and Hommel, U. (2006). Structural basis for the activation of flaviviral NS3 proteases from dengue and West Nile virus. Nature structural & molecular biology, 13 (4): 372-373. Espina, L. M., Valero, N. J., Hernandez, J. M. and Mosquera, J. A. (2003). Increased apoptosis and expression of tumor necrosis factor-α caused by infection of cultured human monocytes with dengue virus. The American journal of tropical medicine and hygiene, 68 (1): 48-53. Esser, M. T., Marchese, R. D., Kierstead, L. S., Tussey, L. G., Wang, F., Chirmule, N. and Washabaugh, M. W. (2003). Memory T cells and vaccines. Vaccine, 21 (5): 419-430. Falgout, B., Chanock, R. and Lai, C. (1989). Proper processing of dengue virus nonstructural glycoprotein NS1 requires the N-terminal hydrophobic signal sequence and the downstream nonstructural protein NS2a. Journal of virology, 63 (5): 1852-1860. Farrar, J. (2008). Clinical features of dengue. SB Halstead. Dengue. London: Imperial College: 171-191. Ferlenghi, I., Clarke, M., Ruttan, T., Allison, S. L., Schalich, J., Heinz, F. X., Harrison, S. C., Rey, F. A. and Fuller, S. D. (2001). Molecular organization of a recombinant subviral particle from tick-borne encephalitis virus. Molecular cell, 7 (3): 593-602. Fernandez-Garcia, M.-D., Mazzon, M., Jacobs, M. and Amara, A. (2009). Pathogenesis of flavivirus infections: using and abusing the host cell. Cell host & microbe, 5 (4): 318-328. Ferreira, G. L. (2012). Global dengue epidemiology trends. Revista do Instituto de Medicina Tropical de São Paulo, 54: 5-6. Filomatori, C. V., Lodeiro, M. F., Alvarez, D. E., Samsa, M. M., Pietrasanta, L. and Gamarnik, A. V. (2006). A 5′ RNA element promotes dengue virus RNA synthesis on a circular genome. Genes & development, 20 (16): 2238-2249. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E. and Mello, C. C. (1998). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. nature, 391 (6669): 806-811. Fischer, L. T. and James, Q. Y. (2004). RNAi, a new therapeutic strategy against viral infection. Cell research, 14 (6): 460-466. Freier, J. E. and Rosen, L. (1988). Vertical transmission of dengue viruses by Aedes mediovittatus. The American journal of tropical medicine and hygiene, 39 (2): 218-222. Fu, Y., Shearing, L. N., Haynes, S., Crewther, P., Tilley, L., Anders, R. F. and Foley, M. (1997). Isolation from phage display libraries of single chain variable fragment antibodies that recognize conformational epitopes in the malaria vaccine candidate, apical membrane antigen-1. Journal of Biological Chemistry, 272 (41): 25678-25684. Garcıá -Montalvo, B. M. A., Medina, F. and Del Angel, R. M. (2004). La protein binds to NS5 and NS3 and to the 5′ and 3′ ends of Dengue 4 virus RNA. Virus research, 102 (2): 141-150. Gebhard, L. G., Filomatori, C. V. and Gamarnik, A. V. (2011). Functional RNA elements in the dengue virus genome. Viruses, 3 (9): 1739-1756.

189

Chapter 6 References

Germi, R., Crance, J.-M., Garin, D., Guimet, J., Lortat-Jacob, H., Ruigrok, R. W., Zarski, J.-P. and Drouet, E. (2002). Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Virology, 292 (1): 162-168. Gibbons, R. V. and Vaughn, D. W. (2002). Dengue: an escalating problem. BMJ: British Medical Journal, 324 (7353): 1563. Goncalvez, A. P., Escalante, A. A., Pujol, F. H., Ludert, J. E., Tovar, D., Salas, R. A. and Liprandi, F. (2002). Diversity and evolution of the envelope gene of dengue virus type 1. Virology, 303 (1): 110-119. Gorbalenya, A. E., Donchenko, A. P., Koonin, E. V. and Blinov, V. M. (1989). N-terminal domains of putative helicases of flavi-and pestiviruses may be serine proteases. Nucleic acids research, 17 (10): 3889-3897. Gu, B., Mason, P., Wang, L., Norton, P., Bourne, N., Moriarty, R., Mehta, A., Despande, M., Shah, R. and Block, T. (2007). Antiviral profiles of novel iminocyclitol compounds against bovine viral diarrhea virus, West Nile virus, dengue virus and hepatitis B virus. Antiviral Chemistry and Chemotherapy, 18 (1): 49-59. Gubler, D. J. (1998). Dengue and dengue hemorrhagic fever. Clinical microbiology reviews, 11 (3): 480-496. Gubler, D. J. (2002). The global emergence/resurgence of arboviral diseases as public health problems. Archives of medical research, 33 (4): 330-342. Gubler, D. J. (2011). Dengue, urbanization and globalization: the unholy trinity of the 21st century. Tropical medicine and health, 39 (4 Suppl): 3. Gubler, D. J., Reed, D., Rosen, L. and Hitchcock Jr, J. (1978). Epidemiologic, clinical, and virologic observations on dengue in the Kingdom of Tonga. The American journal of tropical medicine and hygiene, 27 (3): 581-589. Guirakhoo, F., Heinz, F. X. and Kunz, C. (1989). Epitope model of tick-borne encephalitis virus envelope glycoprotein E: analysis of structural properties, role of carbohydrate side chain, and conformational changes occurring at acidic pH. Virology, 169 (1): 90-99. Guirakhoo, F., Pugachev, K., Zhang, Z., Myers, G., Levenbook, I., Draper, K., Lang, J., Ocran, S., Mitchell, F. and Parsons, M. (2004). Safety and efficacy of chimeric yellow fever-dengue virus tetravalent vaccine formulations in nonhuman primates. Journal of virology, 78 (9): 4761-4775. Guzman, M. G., Deubel, V., Pelegrino, J. L., Rosario, D., Marrero, M., Sariol, C. and Kouri, G. (1995). Partial nucleotide and amino acid sequences of the envelope and the envelope/nonstructural protein-1 gene junction of four dengue-2 virus strains isolated during the 1981 Cuban epidemic. The American journal of tropical medicine and hygiene, 52 (3): 241-246. Guzman, M. G., Halstead, S. B., Artsob, H., Buchy, P., Farrar, J., Gubler, D. J., Hunsperger, E., Kroeger, A., Margolis, H. S. and Martínez, E. (2010). Dengue: a continuing global threat. Nature Reviews Microbiology, 8: S7-S16. Guzman, M. G., Kouri, G., Bravo, J., Calunga, M., Soler, M., Vazquez, S. and Venereo, C. (1984a). Dengue haemorrhagic fever in Cuba. I. Serological confirmation of clinical diagnosis. Transactions of the Royal Society of Tropical Medicine and Hygiene, 78 (2): 235-238. Guzman, M. G., Kouri, G., Bravo, J., Soler, M., Vazquez, S., Santos, M., Villaescusa, R., Basanta, P., Indan, G. and Ballester, J. (1984b). Dengue haemorrhagic fever in Cuba. II. Clinical investigations. Transactions of the Royal Society of Tropical Medicine and Hygiene, 78 (2): 239-241.

190

Chapter 6 References

Guzmán, M. G., Kouri, G. P., Bravo, J., Soler, M., Vazquez, S. and Morier, L. (1990). Dengue hemorrhagic fever in Cuba, 1981: a retrospective seroepidemiologic study. The American journal of tropical medicine and hygiene, 42 (2): 179-184. Halstead, S. (1980). Dengue haemorrhagic fever—a public health problem and a field for research. Bulletin of the World Health Organization, 58 (1): 1. Halstead, S., Chow, J. and Marchette, N. (1973). Immunological enhancement of dengue virus replication. Nature: New biology, 243 (122): 24-26. Halstead, S. and O'rourke, E. (1977). Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody. The Journal of experimental medicine, 146 (1): 201-217. Halstead, S., O'rourke, E. and Allison, A. (1977). Dengue viruses and mononuclear phagocytes. II. Identity of blood and tissue leukocytes supporting in vitro infection. The Journal of experimental medicine, 146 (1): 218-229. Halstead, S. Á., Nimmannitya, S. and Cohen, S. (1970). Observations related to pathogenesis of dengue hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered. The Yale journal of biology and medicine, 42 (5): 311. Halstead, S. B. (2006). Dengue in the Americas and Southeast Asia: do they differ? Revista Panamericana de Salud Publica, 20 (6): 407-415. Halstead, S. B. and Marchette, N. J. (2003). Biologic properties of dengue viruses following serial passage in primary dog kidney cells: studies at the University of Hawaii. The American journal of tropical medicine and hygiene, 69 (6 suppl): 5-11. Hammond, S. M., Bernstein, E., Beach, D. and Hannon, G. J. (2000). An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. nature, 404 (6775): 293- 296. Han, J., Lee, Y., Yeom, K.-H., Kim, Y.-K., Jin, H. and Kim, V. N. (2004). The Drosha-DGCR8 complex in primary microRNA processing. Genes & development, 18 (24): 3016-3027. Harrison, S. C. (2005). Mechanism of membrane fusion by viral envelope proteins. Advances in virus research, 64: 231-261. Heinz, F. X., Mandl, C. W., Holzmann, H., Kunz, C., Harris, B. A., Rey, F. and Harrison, S. C. (1991). The flavivirus envelope protein E: isolation of a soluble form from tick-borne encephalitis virus and its crystallization. Journal of virology, 65 (10): 5579-5583. Heinz, F. X., Stiasny, K. and Allison, S. (2004). The entry machinery of flaviviruses, Springer. Hem, S. L. and Hogenesch, H. (2007). Relationship between physical and chemical properties of aluminum-containing adjuvants and immunopotentiation. Hidari, K. I. and Suzuki, T. (2011). Dengue virus receptor. Tropical medicine and health, 39 (4 Suppl): 37. Hilgard, P. and Stockert, R. (2000). Heparan sulfate proteoglycans initiate dengue virus infection of hepatocytes. Hepatology, 32 (5): 1069-1077. Hirsch, A. J., Medigeshi, G. R., Meyers, H. L., Defilippis, V., Früh, K., Briese, T., Lipkin, W. I. and Nelson, J. A. (2005). The Src family kinase c-Yes is required for maturation of West Nile virus particles. Journal of virology, 79 (18): 11943-11951. Hober, D., Poli, L., Roblin, B., Gestas, P., Chungue, E., Granic, G., Imbert, P., Pecarere, J.-L., Vergez-Pascal, R. and Wattre, P. (1993). Serum levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and interleukin-1 beta (IL-1 beta) in dengue-infected patients. The American journal of tropical medicine and hygiene, 48 (3): 324-331. Holden, K. L., Stein, D. A., Pierson, T. C., Ahmed, A. A., Clyde, K., Iversen, P. L. and Harris, E. (2006). Inhibition of dengue virus translation and RNA synthesis by a morpholino

191

Chapter 6 References

oligomer targeted to the top of the terminal 3′ stem–loop structure. Virology, 344 (2): 439-452. Holmes, E. C. (2003). Patterns of intra-and interhost nonsynonymous variation reveal strong purifying selection in dengue virus. Journal of virology, 77 (20): 11296-11298. Hotta, S. (1952). Experimental studies on dengue I. Isolation, identification and modification of the virus. Journal of Infectious Diseases, 90 (1): 1-9. Hrobowski, Y. M., Garry, R. F. and Michael, S. F. (2005). Peptide inhibitors of dengue virus and West Nile virus infectivity. Virology journal, 2 (1): 49. Huang, Y.-H., Lei, H.-Y., Liu, H.-S., Lin, Y.-S., Liu, C.-C. and Yeh, T.-M. (2000). Dengue virus infects human endothelial cells and induces IL-6 and IL-8 production. The American journal of tropical medicine and hygiene, 63 (1): 71-75. Huang, Y., Chang, B., Lei, H., Liu, H., Wu, H. and Yeh, T. (1997). Antibodies against dengue virus E protein peptide bind to human plasminogen and inhibit plasmin activity. Clinical & Experimental Immunology, 110 (1): 35-40. Huang, Y. H., Lei, H. Y., Liu, H. S., Lin, Y. S., Chen, S. H., Liu, C. C. and Yeh, T. M. (2003). Tissue plasminogen activator induced by dengue virus infection of human endothelial cells. Journal of medical virology, 70 (4): 610-616. Huerta, V., Chinea, G., Fleitas, N., Sarría, M., Sánchez, J., Toledo, P. and Padrón, G. (2008). Characterization of the interaction of domain III of the envelope protein of dengue virus with putative receptors from CHO cells. Virus research, 137 (2): 225-234. Hung, J.-J., Hsieh, M.-T., Young, M.-J., Kao, C.-L., King, C.-C. and Chang, W. (2004). An external loop region of domain III of dengue virus type 2 envelope protein is involved in serotype-specific binding to mosquito but not mammalian cells. Journal of virology, 78 (1): 378-388. Innis, B. L. and Eckels, K. H. (2003). Progress in development of a live-attenuated, tetravalent dengue virus vaccine by the United States Army Medical Research and Materiel Command. The American journal of tropical medicine and hygiene, 69 (6 suppl): 1-4. Innis, B. L., Thirawuth, V. and Hemachudha, C. (1989). Identification of continuous epitopes of the envelope glycoprotein of dengue type 2 virus. The American journal of tropical medicine and hygiene, 40 (6): 676-687. Ishii, K. J. and Akira, S. (2007). Toll or toll-free adjuvant path toward the optimal vaccine development. Journal of clinical immunology, 27 (4): 363-371. Jahan, F. (2011). Dengue fever (DF) in Pakistan. Asia Pac Fam Med, 10 (1): 1. Jindadamrongwech, S., Thepparit, C. and Smith, D. (2004). Identification of GRP 78 (BiP) as a liver cell expressed receptor element for dengue virus serotype 2. Archives of virology, 149 (5): 915-927. Johnson, A. J. and Roehrig, J. T. (1999). New mouse model for dengue virus vaccine testing. Journal of virology, 73 (1): 783-786. Joost Haasnoot, P., Cupac, D. and Berkhout, B. (2003). Inhibition of virus replication by RNA interference. Journal of biomedical science, 10 (6): 607-616. Kanai, R., Kar, K., Anthony, K., Gould, L. H., Ledizet, M., Fikrig, E., Marasco, W. A., Koski, R. A. and Modis, Y. (2006). Crystal structure of West Nile virus envelope glycoprotein reveals viral surface epitopes. Journal of virology, 80 (22): 11000-11008. Kanesa-Thasan, N., Edelman, R., Tacket, C., Wasserman, S., Vaughn, D., Coster, T., Kim-Ahn, G., Dubois, D., Putnak, J. and King, A. (2003). Phase 1 studies of Walter Reed Army Institute of Research candidate attenuated dengue vaccines: selection of safe and immunogenic

192

Chapter 6 References

monovalent vaccines. The American journal of tropical medicine and hygiene, 69 (6 suppl): 17-23. Kaufman, B., Summers, P., Dubois, D., Cohen, W. H. and Gentry, M. (1989). Monoclonal antibodies for dengue virus prM glycoprotein protect mice against lethal dengue infection. DTIC Document. Kautner, I., Robinson, M. J. and Kuhnle, U. (1997). Dengue virus infection: epidemiology, pathogenesis, clinical presentation, diagnosis, and prevention. The Journal of pediatrics, 131 (4): 516-524. Khan, E., Siddiqui, J., Shakoor, S., Mehraj, V., Jamil, B. and Hasan, R. (2007). Dengue outbreak in Karachi, Pakistan, 2006: experience at a tertiary care center. Transactions of the Royal Society of Tropical Medicine and Hygiene, 101 (11): 1114-1119. Khvorova, A., Reynolds, A. and Jayasena, S. D. (2003). Functional siRNAs and miRNAs exhibit strand bias. Cell, 115 (2): 209-216. Kim, D. H. and Rossi, J. J. (2007). Strategies for silencing human disease using RNA interference. Nature Reviews Genetics, 8 (3): 173-184. King, A. D., Nisalak, A., Kalayanarooj, S., Myint, K. S. A., Pattanapanyasat, K., Nimmannitya, S. and Innis, B. L. (1999). B cells are the principal circulating mononuclear cells infected by dengue virus. Southeast Asian journal of tropical medicine and public health, 30 (4): 718- 728. Konishi, E., Kosugi, S. and Imoto, J.-I. (2006). Dengue tetravalent DNA vaccine inducing neutralizing antibody and anamnestic responses to four serotypes in mice. Vaccine, 24 (12): 2200-2207. Konishi, E., Yamaoka, M., Kurane, I. and Mason, P. W. (2000). A DNA vaccine expressing dengue type 2 virus premembrane and envelope genes induces neutralizing antibody and memory B cells in mice. Vaccine, 18 (11): 1133-1139. Kringelum, J. V., Nielsen, M., Padkjær, S. B. and Lund, O. (2013). Structural analysis of B-cell epitopes in antibody: protein complexes. Molecular immunology, 53 (1): 24-34. Krishnan, M. N., Sukumaran, B., Pal, U., Agaisse, H., Murray, J. L., Hodge, T. W. and Fikrig, E. (2007). Rab 5 is required for the cellular entry of dengue and West Nile viruses. Journal of virology, 81 (9): 4881-4885. Kroschewski, H., Lim, S. P., Butcher, R. E., Yap, T. L., Lescar, J., Wright, P. J., Vasudevan, S. G. and Davidson, A. D. (2008). Mutagenesis of the dengue virus type 2 NS5 methyltransferase domain. Journal of Biological Chemistry, 283 (28): 19410-19421. Kuhn, R. J., Zhang, W., Rossmann, M. G., Pletnev, S. V., Corver, J., Lenches, E., Jones, C. T., Mukhopadhyay, S., Chipman, P. R. and Strauss, E. G. (2002). Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell, 108 (5): 717-725. Kümmerer, B. M. and Rice, C. M. (2002). Mutations in the yellow fever virus nonstructural protein NS2A selectively block production of infectious particles. Journal of virology, 76 (10): 4773-4784. Kuno, G., Chang, G.-J. J., Tsuchiya, K. R., Karabatsos, N. and Cropp, C. B. (1998). Phylogeny of the genus Flavivirus. Journal of virology, 72 (1): 73-83. Kurane, I. and Ennis, F. A. (1988). Production of interferon alpha by dengue virus-infected human monocytes. The Journal of general virology, 69: 445-449. Kurane, I., Hebblewaite, D., Brandt, W. E. and Ennis, F. (1984). Lysis of dengue virus-infected cells by natural cell-mediated cytotoxicity and antibody-dependent cell-mediated cytotoxicity. Journal of virology, 52 (1): 223-230.

193

Chapter 6 References

Kurosu, T., Chaichana, P., Yamate, M., Anantapreecha, S. and Ikuta, K. (2007). Secreted complement regulatory protein clusterin interacts with dengue virus nonstructural protein 1. Biochemical and biophysical research communications, 362 (4): 1051-1056. Kuruvilla, J. G., Troyer, R. M., Devi, S. and Akkina, R. (2007). Dengue virus infection and immune response in humanized RAG2−/− γ c−/−(RAG-hu) mice. Virology, 369 (1): 143-152. Kyle, J. L., Beatty, P. R. and Harris, E. (2007). Dengue virus infects macrophages and dendritic cells in a mouse model of infection. Journal of Infectious Diseases, 195 (12): 1808-1817. Kyle, J. L. and Harris, E. (2008). Global spread and persistence of dengue. Annu. Rev. Microbiol., 62: 71-92. Lai, C.-J., Zhao, B., Hori, H. and Bray, M. (1991). Infectious RNA transcribed from stably cloned full-length cDNA of dengue type 4 virus. Proceedings of the National Academy of Sciences, 88 (12): 5139-5143. Lanciotti, R. S., Gubler, D. J. and Trent, D. W. (1997). Molecular evolution and phylogeny of dengue-4 viruses. Journal of General Virology, 78 (9): 2279-2284. Lanciotti, R. S., Lewis, J. G., Gubler, D. J. and Trent, D. W. (1994). Molecular evolution and epidemiology of dengue-3 viruses. Journal of General Virology, 75 (1): 65-76. Leary, J. J., Wittrock, R., Sarisky, R. T., Weinberg, A. and Levin, M. J. (2002). Susceptibilities of herpes simplex viruses to penciclovir and acyclovir in eight cell lines. Antimicrobial agents and chemotherapy, 46 (3): 762-768. Lee, Y., Ahn, C., Han, J., Choi, H., Kim, J., Yim, J., Lee, J., Provost, P., Rådmark, O. and Kim, S. (2003). The nuclear RNase III Drosha initiates microRNA processing. nature, 425 (6956): 415-419. Leng, C.-H., Liu, S.-J., Tsai, J.-P., Li, Y.-S., Chen, M.-Y., Liu, H.-H., Lien, S.-P., Yueh, A., Hsiao, K.-N. and Lai, L.-W. (2009). A novel dengue vaccine candidate that induces cross-neutralizing antibodies and memory immunity. Microbes and Infection, 11 (2): 288-295. Lescar, J., Roussel, A., Wien, M. W., Navaza, J., Fuller, S. D., Wengler, G., Wengler, G. and Rey, F. A. (2001). The fusion glycoprotein shell of Semliki Forest virus: an icosahedral assembly primed for fusogenic activation at endosomal pH. Cell, 105 (1): 137-148. Leung, D., Schroder, K., White, H., Fang, N.-X., Stoermer, M. J., Abbenante, G., Martin, J. L., Young, P. R. and Fairlie, D. P. (2001). Activity of recombinant dengue 2 virus NS3 protease in the presence of a truncated NS2B co-factor, small peptide substrates, and inhibitors. Journal of Biological Chemistry, 276 (49): 45762-45771. Lewis, J. A., Chang, G.-J., Lanciotti, R. S., Kinney, R. M., Mayer, L. W. and Trent, D. W. (1993). Phylogenetic relationships of dengue-2 viruses. Virology, 197 (1): 216-224. Li, L., Lok, S.-M., Yu, I.-M., Zhang, Y., Kuhn, R. J., Chen, J. and Rossmann, M. G. (2008). The flavivirus precursor membrane-envelope protein complex: structure and maturation. Science, 319 (5871): 1830-1834. Liao, M. and Kielian, M. (2005). Domain III from class II fusion proteins functions as a dominant- negative inhibitor of virus membrane fusion. The Journal of cell biology, 171 (1): 111- 120. Libraty, D. H., Endy, T. P., Houng, H.-S. H., Green, S., Kalayanarooj, S., Suntayakorn, S., Chansiriwongs, W., Vaughn, D. W., Nisalak, A. and Ennis, F. A. (2002). Differing influences of virus burden and immune activation on disease severity in secondary dengue-3 virus infections. Journal of Infectious Diseases, 185 (9): 1213-1221. Lim, S. P., Sonntag, L. S., Noble, C., Nilar, S. H., Ng, R. H., Zou, G., Monaghan, P., Chung, K. Y., Dong, H. and Liu, B. (2011). Small molecule inhibitors that selectively block dengue virus methyltransferase. Journal of Biological Chemistry, 286 (8): 6233-6240.

194

Chapter 6 References

Lin, C.-F., Chiu, S.-C., Hsiao, Y.-L., Wan, S.-W., Lei, H.-Y., Shiau, A.-L., Liu, H.-S., Yeh, T.-M., Chen, S.-H. and Liu, C.-C. (2005). Expression of cytokine, chemokine, and adhesion molecules during endothelial cell activation induced by antibodies against dengue virus nonstructural protein 1. The Journal of Immunology, 174 (1): 395-403. Lin, C.-F., Wan, S.-W., Chen, M.-C., Lin, S.-C., Cheng, C.-C., Chiu, S.-C., Hsiao, Y.-L., Lei, H.-Y., Liu, H.-S. and Yeh, T.-M. (2008a). Liver injury caused by antibodies against dengue virus nonstructural protein 1 in a murine model. Laboratory Investigation, 88 (10): 1079-1089. Lin, H.-E., Tsai, W.-Y., Liu, I.-J., Li, P.-C., Liao, M.-Y., Tsai, J.-J., Wu, Y.-C., Lai, C.-Y., Lu, C.-H. and Huang, J.-H. (2012). Analysis of epitopes on dengue virus envelope protein recognized by monoclonal antibodies and polyclonal human sera by a high throughput assay. PLoS Negl Trop Dis, 6 (1): e1447-e1447. Lin, H. H., Ray, S., Tongchusak, S., Reinherz, E. L. and Brusic, V. (2008b). Evaluation of MHC class I peptide binding prediction servers: applications for vaccine research. BMC immunology, 9 (1): 8. Lin, Y.-J. and Wu, S.-C. (2005). Histidine at residue 99 and the transmembrane region of the precursor membrane prM protein are important for the prM-E heterodimeric complex formation of Japanese encephalitis virus. Journal of virology, 79 (13): 8535-8544. Lindenbach, B. D. and Rice, C. (2001). Flaviviridae: the viruses and their replication. Fields virology, 1: 991-1041. Littaua, R., Kurane, I. and Ennis, F. A. (1990). Human IgG Fc receptor II mediates antibody- dependent enhancement of dengue virus infection. The Journal of Immunology, 144 (8): 3183-3186. Liu, J., Carmell, M. A., Rivas, F. V., Marsden, C. G., Thomson, J. M., Song, J.-J., Hammond, S. M., Joshua-Tor, L. and Hannon, G. J. (2004). Argonaute2 is the catalytic engine of mammalian RNAi. Science, 305 (5689): 1437-1441. Liu, W. J., Chen, H. B. and Khromykh, A. A. (2003). Molecular and functional analyses of Kunjin virus infectious cDNA clones demonstrate the essential roles for NS2A in virus assembly and for a nonconservative residue in NS3 in RNA replication. Journal of virology, 77 (14): 7804-7813. Livingston, P. G., Kurane, I., Dai, L.-C., Okamoto, Y., Lai, C.-J., Men, R., Karaki, S., Takiguchi, M. and Ennis, F. A. (1995). Dengue virus-specific, HLA-B35-restricted, human CD8+ cytotoxic T lymphocyte (CTL) clones. Recognition of NS3 amino acids 500 to 508 by CTL clones of two different serotype specificities. The Journal of Immunology, 154 (3): 1287- 1295. Lobigs, M. (1993). Flavivirus premembrane protein cleavage and spike heterodimer secretion require the function of the viral proteinase NS3. Proceedings of the National Academy of Sciences, 90 (13): 6218-6222. Lozach, P.-Y., Burleigh, L., Staropoli, I., Navarro-Sanchez, E., Harriague, J., Virelizier, J.-L., Rey, F. A., Desprès, P., Arenzana-Seisdedos, F. and Amara, A. (2005). Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals. Journal of Biological Chemistry, 280 (25): 23698-23708. Luplerdlop, N., Missé, D., Bray, D., Deleuze, V., Gonzalez, J. P., Leardkamolkarn, V., Yssel, H. and Veas, F. (2006). Dengue‐virus‐infected dendritic cells trigger vascular leakage through metalloproteinase overproduction. EMBO reports, 7 (11): 1176-1181. Luzhkov, V. B., Selisko, B., Nordqvist, A., Peyrane, F., Decroly, E., Alvarez, K., Karlen, A., Canard, B. and Åqvist, J. (2007). Virtual screening and bioassay study of novel inhibitors for

195

Chapter 6 References

dengue virus mRNA cap (nucleoside-2′ O)-methyltransferase. Bioorganic & medicinal chemistry, 15 (24): 7795-7802. Ma, L., Jones, C. T., Groesch, T. D., Kuhn, R. J. and Post, C. B. (2004). Solution structure of dengue virus capsid protein reveals another fold. Proceedings of the National Academy of Sciences of the United States of America, 101 (10): 3414-3419. Mackenzie, J. M., Jones, M. K. and Young, P. R. (1996). Immunolocalization of the dengue virus nonstructural glycoprotein NS1 suggests a role in viral RNA replication. Virology, 220 (1): 232-240. Mackenzie, J. M., Khromykh, A. A., Jones, M. K. and Westaway, E. G. (1998). Subcellular localization and some biochemical properties of the flavivirus Kunjin nonstructural proteins NS2A and NS4A. Virology, 245 (2): 203-215. Mackenzie, J. M. and Westaway, E. G. (2001). Assembly and maturation of the flavivirus Kunjin virus appear to occur in the rough endoplasmic reticulum and along the secretory pathway, respectively. Journal of virology, 75 (22): 10787-10799. Mahalingam, S., Herring, B. L. and Halstead, S. B. (2013). Call to action for dengue vaccine failure. Emerging infectious diseases, 19 (8): 1335. Mahmood, N., Rana, M. Y., Qureshi, Z., Mujtaba, G. and Shaukat, U. (2012). Prevalence and molecular characterization of dengue viruses serotypes in 2010 epidemic. The American journal of the medical sciences, 343 (1): 61-64. Manzano, M., Reichert, E. D., Polo, S., Falgout, B., Kasprzak, W., Shapiro, B. A. and Padmanabhan, R. (2011). Identification of cis-acting elements in the 3′-untranslated region of the dengue virus type 2 RNA that modulate translation and replication. Journal of Biological Chemistry, 286 (25): 22521-22534. Markoff, L. (2003). 5′-and 3′-noncoding regions in flavivirus RNA. Advances in virus research, 59: 177-228. Markoff, L. J., Innis, B. L., Houghten, R. and Henchal, L. S. (1991). Development of cross-reactive antibodies to plasminogen during the immune response to dengue virus infection. Journal of Infectious Diseases, 164 (2): 294-301. Marovich, M., Grouard-Vogel, G., Louder, M., Eller, M., Sun, W., Wu, S.-J., Putvatana, R., Murphy, G., Tassaneetrithep, B. and Burgess, T. Year. Human dendritic cells as targets of dengue virus infection. In: Journal of Investigative Dermatology Symposium Proceedings, 2001. Nature Publishing Group, 219-224. Mcarthur, J. H., Durbin, A. P., Marron, J. A., Wanionek, K. A., Thumar, B., Pierro, D. J., Schmidt, A. C., Blaney, J. E., Murphy, B. R. and Whitehead, S. S. (2008). Phase I clinical evaluation of rDEN4Δ30-200,201: a live attenuated dengue 4 vaccine candidate designed for decreased hepatotoxicity. The American journal of tropical medicine and hygiene, 79 (5): 678-684. Megret, F., Hugnot, J., Falconar, A., Gentry, M., Morens, D., Murray, J., Schlesinger, J., Wright, P., Young, P. and Van Regenmortel, M. (1992). Use of recombinant fusion proteins and monoclonal antibodies to define linear and discontinuous antigenic sites on the dengue virus envelope glycoprotein. Virology, 187 (2): 480-491. Meister, G., Landthaler, M., Patkaniowska, A., Dorsett, Y., Teng, G. and Tuschl, T. (2004). Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Molecular cell, 15 (2): 185-197. Meister, G. and Tuschl, T. (2004). Mechanisms of gene silencing by double-stranded RNA. Nature, 431 (7006): 343-349.

196

Chapter 6 References

Mello, C. C. and Conte, D. (2004). Revealing the world of RNA interference. Nature, 431 (7006): 338-342. Men, R., Wyatt, L., Tokimatsu, I., Arakaki, S., Shameem, G., Elkins, R., Chanock, R., Moss, B. and Lai, C.-J. (2000). 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, 18 (27): 3113-3122. Mendoza, M. Y., Salas-Benito, J. S., Lanz-Mendoza, H., Hernández-Martínez, S. and Del Angel, R. M. (2002). A putative receptor for dengue virus in mosquito tissues: localization of a 45- kDa glycoprotein. The American journal of tropical medicine and hygiene, 67 (1): 76-84. Mercado-Curiel, R. F., Black, W. C. and De L Muñoz, M. (2008). A dengue receptor as possible genetic marker of vector competence in Aedes aegypti. BMC microbiology, 8 (1): 118. Messer, W. B., Gubler, D. J., Harris, E., Sivananthan, K. and De Silva, A. M. (2003). Emergence and global spread of a dengue serotype 3, subtype III virus. Emerging infectious diseases, 9 (7): 800. Migliaccio, G., Tomassini, J. E., Carroll, S. S., Tomei, L., Altamura, S., Bhat, B., Bartholomew, L., Bosserman, M. R., Ceccacci, A. and Colwell, L. F. (2003). Characterization of resistance to non-obligate chain-terminating ribonucleoside analogs that inhibit replication in vitro. Journal of Biological Chemistry, 278 (49): 49164-49170. Miller, J. L., De Wet, B., Martinez-Pomares, L., Radcliffe, C. M., Dwek, R. A., Rudd, P. M. and Gordon, S. (2008). The mannose receptor mediates dengue virus infection of macrophages. PLoS Pathog, 4 (2): e17. Miller, S., Kastner, S., Krijnse-Locker, J., Bühler, S. and Bartenschlager, R. (2007). The non- structural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner. Journal of Biological Chemistry, 282 (12): 8873-8882. Miller, S., Sparacio, S. and Bartenschlager, R. (2006). Subcellular localization and membrane topology of the dengue virus type 2 non-structural protein 4B. Journal of Biological Chemistry, 281 (13): 8854-8863. Modis, Y., Ogata, S., Clements, D. and Harrison, S. C. (2003). A ligand-binding pocket in the dengue virus envelope glycoprotein. Proceedings of the National Academy of Sciences, 100 (12): 6986-6991. Modis, Y., Ogata, S., Clements, D. and Harrison, S. C. (2004). Structure of the dengue virus envelope protein after membrane fusion. Nature, 427 (6972): 313-319. Molina-Cruz, A., Gupta, L., Richardson, J., Bennett, K., Black, W. and Barillas-Mury, C. (2005). Effect of mosquito midgut trypsin activity on dengue-2 virus infection and dissemination in Aedes aegypti. The American journal of tropical medicine and hygiene, 72 (5): 631- 637. Monath, T. P. (1994). Dengue: the risk to developed and developing countries. Proceedings of the National Academy of Sciences, 91 (7): 2395-2400. Mongkolsapaya, J., Duangchinda, T., Dejnirattisai, W., Vasanawathana, S., Avirutnan, P., Jairungsri, A., Khemnu, N., Tangthawornchaikul, N., Chotiyarnwong, P. and Sae-Jang, K. (2006). T cell responses in dengue hemorrhagic fever: are cross-reactive T cells suboptimal? The Journal of Immunology, 176 (6): 3821-3829. Montgomery, M. K., Xu, S. and Fire, A. (1998). RNA as a target of double-stranded RNA- mediated genetic interference in Caenorhabditis elegans. Proceedings of the National Academy of Sciences, 95 (26): 15502-15507.

197

Chapter 6 References

Mueller, N. H., Pattabiraman, N., Ansarah-Sobrinho, C., Viswanathan, P., Pierson, T. C. and Padmanabhan, R. (2008). Identification and biochemical characterization of small- molecule inhibitors of West Nile virus serine protease by a high-throughput screen. Antimicrobial agents and chemotherapy, 52 (9): 3385-3393. Mueller, N. H., Yon, C., Ganesh, V. K. and Padmanabhan, R. (2007). Characterization of the West Nile virus protease substrate specificity and inhibitors. The international journal of biochemistry & cell biology, 39 (3): 606-614. Muñoz-Jordán, J. L., Sánchez-Burgos, G. G., Laurent-Rolle, M. and García-Sastre, A. (2003). Inhibition of interferon signaling by dengue virus. Proceedings of the National Academy of Sciences, 100 (24): 14333-14338. Murray, N. E. A., Quam, M. B. and Wilder-Smith, A. (2013). Epidemiology of dengue: past, present and future prospects. Clinical epidemiology, 5: 299. Muzzi, A., Masignani, V. and Rappuoli, R. (2007). The pan-genome: towards a knowledge-based discovery of novel targets for vaccines and antibacterials. Drug discovery today, 12 (11): 429-439. Navarro‐Sanchez, E., Altmeyer, R., Amara, A., Schwartz, O., Fieschi, F., Virelizier, J. L., Arenzana‐Seisdedos, F. and Desprès, P. (2003). Dendritic‐cell‐specific ICAM3‐grabbing non‐integrin is essential for the productive infection of human dendritic cells by mosquito‐cell‐derived dengue viruses. EMBO reports, 4 (7): 723-728. Noble, C. G., Chen, Y.-L., Dong, H., Gu, F., Lim, S. P., Schul, W., Wang, Q.-Y. and Shi, P.-Y. (2010). Strategies for development of dengue virus inhibitors. Antiviral research, 85 (3): 450- 462. Noisakran, S., Sengsai, S., Thongboonkerd, V., Kanlaya, R., Sinchaikul, S., Chen, S.-T., Puttikhunt, C., Kasinrerk, W., Limjindaporn, T. and Wongwiwat, W. (2008). Identification of human hnRNP C1/C2 as a dengue virus NS1-interacting protein. Biochemical and biophysical research communications, 372 (1): 67-72. Nowak, T. and Wengler, G. (1987). Analysis of disulfides present in the membrane proteins of the West Nile flavivirus. Virology, 156 (1): 127-137. Palmer, D. R., Sun, P., Celluzzi, C., Bisbing, J., Pang, S., Sun, W., Marovich, M. A. and Burgess, T. (2005). Differential effects of dengue virus on infected and bystander dendritic cells. Journal of virology, 79 (4): 2432-2439. Pastor-Anglada, M., Cano-Soldado, P., Molina-Arcas, M., Lostao, M. P., Larráyoz, I., Martínez- Picado, J. and Casado, F. J. (2005). Cell entry and export of nucleoside analogues. Virus research, 107 (2): 151-164. Paul, R. E., Patel, A. Y., Mirza, S., Fisher-Hoch, S. P. and Luby, S. P. (1998). Expansion of epidemic dengue viral infections to Pakistan. International journal of infectious diseases, 2 (4): 197-201. Podvinec, M., Lim, S. P., Schmidt, T., Scarsi, M., Wen, D., Sonntag, L.-S., Sanschagrin, P., Shenkin, P. S. and Schwede, T. (2010). Novel inhibitors of dengue virus methyltransferase: discovery by in vitro-driven virtual screening on a desktop computer grid. Journal of medicinal chemistry, 53 (4): 1483-1495. Pollock, S., Antrobus, R., Newton, L., Kampa, B., Rossa, J., Latham, S., Nichita, N. B., Dwek, R. A. and Zitzmann, N. (2010). Uptake and trafficking of liposomes to the endoplasmic reticulum. The FASEB Journal, 24 (6): 1866-1878. Polyreactive, D. V. A. (2011). Dengue virus activates polyreactive, natural IgG B cells after primary and secondary infection.

198

Chapter 6 References

Poovorawan, Y., Hutagalung, Y., Chongsrisawat, V., Boudville, I. and Bock, H. L. (2006). Dengue virus infection: a major cause of acute hepatic failure in Thai children. Annals of Tropical Paediatrics: International Child Health, 26 (1): 17-23. Pulendran, B. and Ahmed, R. (2006). Translating innate immunity into immunological memory: implications for vaccine development. Cell, 124 (4): 849-863. Putnak, J. R., Coller, B.-A., Voss, G., Vaughn, D. W., Clements, D., Peters, I., Bignami, G., Houng, H.-S., Chen, R. C.-M. and Barvir, D. A. (2005). An evaluation of dengue type-2 inactivated, recombinant subunit, and live-attenuated vaccine candidates in the rhesus macaque model. Vaccine, 23 (35): 4442-4452. Ramanathan, M. P., Kuo, Y.-C., Selling, B. H., Li, Q., Sardesai, N. Y., Kim, J. J. and Weiner, D. B. (2009). Development of a novel DNA SynCon™ tetravalent dengue vaccine that elicits immune responses against four serotypes. Vaccine, 27 (46): 6444-6453. Range, H. (2008). Dengue: Overview and History. Dengue, 5: 1. Raut, C., Deolankar, R., Kolhapure, R. and Goverdhan, M. (1996). SUSCEPTIBILITY OF LABORATORY-BRED RODENTS.. TO THE EXPERIMENTAL INFECnON WITH DENGUE VIRUS TYPE 2. Acta vil> gica, 40 (143): 146. Raviprakash, K., Wang, D., Ewing, D., Holman, D. H., Block, K., Woraratanadharm, J., Chen, L., Hayes, C., Dong, J. Y. and Porter, K. (2008). A tetravalent dengue vaccine based on a complex adenovirus vector provides significant protection in rhesus monkeys against all four serotypes of dengue virus. Journal of virology, 82 (14): 6927-6934. Ray, D., Shah, A., Tilgner, M., Guo, Y., Zhao, Y., Dong, H., Deas, T. S., Zhou, Y., Li, H. and Shi, P.-Y. (2006). West Nile virus 5′-cap structure is formed by sequential guanine N-7 and ribose 2′-O methylations by nonstructural protein 5. Journal of virology, 80 (17): 8362-8370. Rees, C. R., Costin, J. M., Fink, R. C., Mcmichael, M., Fontaine, K. A., Isern, S. and Michael, S. F. (2008). In vitro inhibition of dengue virus entry by p-sulfoxy-cinnamic acid and structurally related combinatorial chemistries. Antiviral research, 80 (2): 135-142. Rey, F. A., Heinz, F. X., Mandl, C., Kunz, C. and Harrison, S. C. (1995). The envelope glycoprotein from tick-borne encephalitis virus at 2 Å resolution. Rice, C. M., Lenches, E. M., Shin, S., Sheets, R. and Strauss, J. (1985). Nucleotide sequence of yellow fever virus: implications for flavivirus gene expression and evolution. Science, 229 (4715): 726-733. Richter, M. K., Da Silva Voorham, J. M., Pedraza, S. T., Hoornweg, T. E., Van De Pol, D. P., Rodenhuis-Zybert, I. A., Wilschut, J. and Smit, J. M. (2014). Immature Dengue Virus Is Infectious in Human Immature Dendritic Cells via Interaction with the Receptor Molecule DC-SIGN. Rico-Hesse, R. (1990). Molecular evolution and distribution of dengue viruses type 1 and 2 in nature. Virology, 174 (2): 479-493. Rico-Hesse, R., Harrison, L. M., Salas, R. A., Tovar, D., Nisalak, A., Ramos, C., Boshell, J., De Mesa, M. T. R., Nogueira, R. M. and Da Rosa, A. T. (1997). Origins of dengue type 2 viruses associated with increased pathogenicity in the Americas. Virology, 230 (2): 244-251. Rigau-Pérez, J. G., Clark, G. G., Gubler, D. J., Reiter, P., Sanders, E. J. and Vorndam, A. V. (1998). Dengue and dengue haemorrhagic fever. The Lancet, 352 (9132): 971-977. Roby, J. A., Pijlman, G. P., Wilusz, J. and Khromykh, A. A. (2014). Noncoding subgenomic flavivirus RNA: multiple functions in West Nile virus pathogenesis and modulation of host responses. Viruses, 6 (2): 404-427. Roehrig, J. T. (2003). Antigenic structure of flavivirus proteins. Advances in virus research, 59: 141-175.

199

Chapter 6 References

Roehrig, J. T., Bolin, R. A. and Kelly, R. G. (1998). Monoclonal antibody mapping of the envelope glycoprotein of the dengue 2 virus, Jamaica. Virology, 246 (2): 317-328. Rothman, A. L. (2004). Dengue: defining protective versus pathologic immunity. Journal of Clinical Investigation, 113 (7): 946. Sabchareon, A., Lang, J., Chanthavanich, P., Yoksan, S., Forrat, R., Attanath, P., Sirivichayakul, C., Pengsaa, K., Pojjaroen-Anant, C. and Chambonneau, L. (2004). Safety and immunogenicity of a three dose regimen of two tetravalent live-attenuated dengue vaccines in five-to twelve-year-old Thai children. The Pediatric infectious disease journal, 23 (2): 99-109. Sabchareon, A., Lang, J., Chanthavanich, P., Yoksan, S., Forrat, R., Attanath, P., Sirivichayakul, C., Pengsaa, K., Pojjaroen-Anant, C. and Chokejindachai, W. (2002). Safety and immunogenicity of tetravalent live-attenuated dengue vaccines in Thai adult volunteers: role of serotype concentration, ratio, and multiple doses. The American journal of tropical medicine and hygiene, 66 (3): 264-272. Sabchareon, A., Wallace, D., Sirivichayakul, C., Limkittikul, K., Chanthavanich, P., Suvannadabba, S., Jiwariyavej, V., Dulyachai, W., Pengsaa, K. and Wartel, T. A. (2012). Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. The Lancet, 380 (9853): 1559- 1567. Sabin, A. B. (1952). Research on dengue during World War II. The American journal of tropical medicine and hygiene, 1 (1): 30-50. Sakoonwatanyoo, P., Boonsanay, V. and Smith, D. R. (2006). Growth and production of the dengue virus in C6/36 cells and identification of a laminin-binding protein as a candidate serotype 3 and 4 receptor protein. Intervirology, 49 (3): 161-172. Salazar, M. I., Richardson, J. H., Sánchez-Vargas, I., Olson, K. E. and Beaty, B. J. (2007). Dengue virus type 2: replication and tropisms in orally infected Aedes aegypti mosquitoes. BMC microbiology, 7 (1): 9. Sall, A. A., Faye, O., Diallo, M., Firth, C., Kitchen, A. and Holmes, E. C. (2010). Yellow fever virus exhibits slower evolutionary dynamics than dengue virus. Journal of virology, 84 (2): 765-772. Sánchez-Vargas, I., Scott, J. C., Poole-Smith, B. K., Franz, A. W., Barbosa-Solomieu, V., Wilusz, J., Olson, K. E. and Blair, C. D. (2009). Dengue virus type 2 infections of Aedes aegypti are modulated by the mosquito’s RNA interference pathway. PLoS Pathog, 5 (2): e1000299. Sanchez-Vargas, I., Travanty, E. A., Keene, K. M., Franz, A. W., Beaty, B. J., Blair, C. D. and Olson, K. E. (2004). RNA interference, arthropod-borne viruses, and mosquitoes. Virus research, 102 (1): 65-74. Schmid, M. A., Diamond, M. S. and Harris, E. (2014). Dendritic cells in dengue virus infection: targets of virus replication and mediators of immunity. Frontiers in immunology, 5. Schwarz, D. S., Hutvágner, G., Du, T., Xu, Z., Aronin, N. and Zamore, P. D. (2003). Asymmetry in the assembly of the RNAi enzyme complex. Cell, 115 (2): 199-208. Scott, R. M., Nisalak, A., Seridhoranakul, S. and Nimmannitya, S. (1980). Isolation of dengue viruses from peripheral blood leukocytes of patients with hemorrhagic fever. Journal of Infectious Diseases, 141 (1): 1-6. Screaton, G. and Mongkolsapaya, J. Year. T cell responses and dengue haemorrhagic fever. In: Novartis Foundation symposium, 2006. Chichester; New York; John Wiley; 1999, 164.

200

Chapter 6 References

Sessions, O. M., Barrows, N. J., Souza-Neto, J. A., Robinson, T. J., Hershey, C. L., Rodgers, M. A., Ramirez, J. L., Dimopoulos, G., Yang, P. L. and Pearson, J. L. (2009). Discovery of insect and human dengue virus host factors. Nature, 458 (7241): 1047-1050. Sette, A., Livingston, B., Mckinney, D., Appella, E., Fikes, J., Sidney, J., Newman, M. and Chesnut, R. (2001). The development of multi-epitope vaccines: epitope identification, vaccine design and clinical evaluation. Biologicals, 29 (3): 271-276. Shafee, N. and Abubakar, S. (2003). Dengue virus type 2 NS3 protease and NS2B-NS3 protease precursor induce apoptosis. Journal of general virology, 84 (8): 2191-2195. Shaio, M.-F., Chang, F.-Y. and Hou, S.-C. (1992). Complement pathway activity in serum from patients with classical dengue fever. Transactions of the Royal Society of Tropical Medicine and Hygiene, 86 (6): 672-675. Shastri, N., Schwab, S. and Serwold, T. (2002). Producing nature's gene-chips: the generation of peptides for display by MHC class I molecules. Annual review of immunology, 20 (1): 463-493. Shepard, D. S., Coudeville, L., Halasa, Y. A., Zambrano, B. and Dayan, G. H. (2011). Economic impact of dengue illness in the Americas. The American journal of tropical medicine and hygiene, 84 (2): 200-207. Shepard, D. S., Undurraga, E. A. and Halasa, Y. A. (2013). Economic and disease burden of dengue in Southeast Asia. PLoS Negl Trop Dis, 7 (2): e2055. Shresta, S., Sharar, K. L., Prigozhin, D. M., Beatty, P. R. and Harris, E. (2006). Murine model for dengue virus-induced lethal disease with increased vascular permeability. Journal of virology, 80 (20): 10208-10217. Simmons, C. P., Dong, T., Chau, N. V., Dung, N. T. P., Chau, T. N. B., Dung, N. T., Hien, T. T., Rowland-Jones, S. and Farrar, J. (2005). Early T-cell responses to dengue virus epitopes in Vietnamese adults with secondary dengue virus infections. Journal of virology, 79 (9): 5665-5675. Simmons, J. S., St John, J. H. and Reynolds, F. H. (1931). Experimental studies of dengue. Monogr. Bur. Sci. Philipp. Is., (29). Simmons, M., Burgess, T., Lynch, J. and Putnak, R. (2010). Protection against dengue virus by non-replicating and live attenuated vaccines used together in a prime boost vaccination strategy. Virology, 396 (2): 280-288. Sirigulpanit, W., Kinney, R. M. and Leardkamolkarn, V. (2007). Substitution or deletion mutations between nt 54 and 70 in the 5′ non-coding region of dengue type 2 virus produce variable effects on virus viability. Journal of general virology, 88 (6): 1748-1752. Sloan-Lancaster, J. and Allen, P. M. (1996). Altered peptide ligand-induced partial T cell activation: molecular mechanisms and role in T cell biology. Annual review of immunology, 14 (1): 1-27. Soriano, V., Madejon, A., Vispo, E., Labarga, P., Garcia-Samaniego, J., Martin-Carbonero, L., Sheldon, J., Bottecchia, M., Tuma, P. and Barreiro, P. (2008). Emerging drugs for hepatitis C. Souza, D. G., Fagundes, C. T., Sousa, L. P., Amaral, F. A., Souza, R. S., Souza, A. L., Kroon, E. G., Sachs, D., Cunha, F. Q. and Bukin, E. (2009). Essential role of platelet-activating factor receptor in the pathogenesis of Dengue virus infection. Proceedings of the National Academy of Sciences, 106 (33): 14138-14143. Srikiatkhachorn, A., Ajariyakhajorn, C., Endy, T. P., Kalayanarooj, S., Libraty, D. H., Green, S., Ennis, F. A. and Rothman, A. L. (2007). Virus-induced decline in soluble vascular

201

Chapter 6 References

endothelial growth receptor 2 is associated with plasma leakage in dengue hemorrhagic Fever. Journal of virology, 81 (4): 1592-1600. Stadler, K., Allison, S. L., Schalich, J. and Heinz, F. X. (1997). Proteolytic activation of tick-borne encephalitis virus by furin. Journal of Virology, 71 (11): 8475-8481. Steidel, M., Fragnoud, R., Guillotte, M., Roesch, C., Michel, S., Meunier, T., Paranhos‐Baccalà, G., Gervasi, G. and Bedin, F. (2012). Nonstructural protein NS1 immunodominant epitope detected specifically in dengue virus infected material by a SELDI‐TOF/MS based assay. Journal of medical virology, 84 (3): 490-499. Steinhauer, D. A., Domingo, E. and Holland, J. J. (1992). Lack of evidence for proofreading mechanisms associated with an RNA virus polymerase. Gene, 122 (2): 281-288. Stiasny, K., Allison, S. L., Marchler-Bauer, A., Kunz, C. and Heinz, F. X. (1996). Structural requirements for low-pH-induced rearrangements in the envelope glycoprotein of tick- borne encephalitis virus. Journal of virology, 70 (11): 8142-8147. Subramanya, S., Kim, S.-S., Abraham, S., Yao, J., Kumar, M., Kumar, P., Haridas, V., Lee, S.-K., Shultz, L. D. and Greiner, D. (2010). Targeted delivery of small interfering RNA to human dendritic cells to suppress dengue virus infection and associated proinflammatory cytokine production. Journal of virology, 84 (5): 2490-2501. Sukri, N. C., Laras, K., Wandra, T., Didi, S., Larasati, R. P., Rachdyatmaka, J. R., Osok, S., Tjia, P., Saragih, J. M. and Hartati, S. (2003). Transmission of epidemic dengue hemorrhagic fever in easternmost Indonesia. The American journal of tropical medicine and hygiene, 68 (5): 529-535. Sun, E., Zhao, J., Liu, N., Yang, T., Xu, Q., Qin, Y., Bu, Z., Yang, Y., Lunt, R. A. and Wang, L. (2012). Comprehensive mapping of common immunodominant epitopes in the West Nile virus nonstructural protein 1 recognized by avian antibody responses. PloS one, 7 (2): e31434. Sun, P. and Kochel, T. J. (2013). The battle between infection and host immune responses of dengue virus and its implication in dengue disease pathogenesis. The Scientific World Journal, 2013. Sun, W., Edelman, R., Kanesa-Thasan, N., Eckels, K. H., Putnak, J. R., King, A. D., Houng, H.-S., Tang, D., Scherer, J. M. and Hoke, C. H. (2003). Vaccination of human volunteers with monovalent and tetravalent live-attenuated dengue vaccine candidates. The American journal of tropical medicine and hygiene, 69 (6 suppl): 24-31. Svitkin, Y. V., Lyapustin, V. N., Lashkevich, V. A. and Agol, V. I. (1984). Differences between translation products of tick-borne encephalitis virus RNA in cell-free systems from Krebs-2 cells and rabbit reticulocytes: involvement of membranes in the processing of nascent precursors of flavivirus structural proteins. Virology, 135 (2): 536-541. Sweredoski, M. J. and Baldi, P. (2009). COBEpro: a novel system for predicting continuous B-cell epitopes. Protein Engineering Design and Selection, 22 (3): 113-120. Takahashi, H., Merli, S., Putney, S. D., Houghten, R., Moss, B., Germain, R. N. and Berzofsky, J. A. (1989). A single amino acid interchange yields reciprocal CTL specificities for HIV-1 gp160. Science, 246 (4926): 118-121. Tang, J. W., Khanani, M. R., Zubairi, A. M., Lam, W. Y., Lai, F., Hashmi, K., Hussain, A., Jamal, S. and Chan, P. K. (2008). A wide spectrum of dengue IgM and PCR positivity post‐onset of illness found in a large dengue 3 outbreak in Pakistan. Journal of medical virology, 80 (12): 2113-2121. Tassaneetrithep, B., Burgess, T. H., Granelli-Piperno, A., Trumpfheller, C., Finke, J., Sun, W., Eller, M. A., Pattanapanyasat, K., Sarasombath, S. and Birx, D. L. (2003). DC-SIGN (CD209)

202

Chapter 6 References

mediates dengue virus infection of human dendritic cells. The Journal of experimental medicine, 197 (7): 823-829. Thepparit, C. and Smith, D. R. (2004). 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. Journal of virology, 78 (22): 12647-12656. Tilgner, M., Deas, T. S. and Shi, P.-Y. (2005). The flavivirus-conserved penta-nucleotide in the 3′ stem-loop of the West Nile virus genome requires a specific sequence and structure for RNA synthesis, but not for viral translation. Virology, 331 (2): 375-386. Tomlinson, S. M., Malmstrom, R. D., Russo, A., Mueller, N., Pang, Y.-P. and Watowich, S. J. (2009). Structure-based discovery of dengue virus protease inhibitors. Antiviral research, 82 (3): 110-114. Trent, D., Shin, J., Hombach, J., Knezevic, I., Minor, P. and Group, W. W. (2010). WHO Working Group on technical specifications for manufacture and evaluation of dengue vaccines, Geneva, Switzerland, 11–12 May 2009. Vaccine, 28 (52): 8246-8255. Twiddy, S. S., Farrar, J. J., Chau, N. V., Wills, B., Gould, E. A., Gritsun, T., Lloyd, G. and Holmes, E. C. (2002). Phylogenetic relationships and differential selection pressures among genotypes of dengue-2 virus. Virology, 298 (1): 63-72. Uchil, P. D. and Satchidanandam, V. (2003). Architecture of the flaviviral replication complex protease, nuclease, and detergents reveal encasement within double-layered membrane compartments. Journal of Biological Chemistry, 278 (27): 24388-24398. Umareddy, I., Chao, A., Sampath, A., Gu, F. and Vasudevan, S. G. (2006). Dengue virus NS4B interacts with NS3 and dissociates it from single-stranded RNA. Journal of general virology, 87 (9): 2605-2614. Unkeless, J. (1989). Function and heterogeneity of human Fc receptors for immunoglobulin G. Journal of Clinical Investigation, 83 (2): 355. Upanan, S., Kuadkitkan, A. and Smith, D. R. (2008). Identification of dengue virus binding proteins using affinity chromatography. Journal of virological methods, 151 (2): 325-328. Valle, R. P. and Falgout, B. (1998). Mutagenesis of the NS3 protease of dengue virus type 2. Journal of virology, 72 (1): 624-632. Vasilakis, N. and Weaver, S. C. (2008). The history and evolution of human dengue emergence. Advances in virus research, 72: 1-76. Vaughn, D. W., Green, S., Kalayanarooj, S., Innis, B. L., Nimmannitya, S., Suntayakorn, S., Endy, T. P., Raengsakulrach, B., Rothman, A. L. and Ennis, F. A. (2000). Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. Journal of Infectious Diseases, 181 (1): 2-9. Vázquez, S., Guzmán, M. a. G., Guillen, G., Chinea, G., Pérez, A. B., Pupo, M., Rodriguez, R., Reyes, O., Garay, H. E. and Delgado, I. (2002). Immune response to synthetic peptides of dengue prM protein. Vaccine, 20 (13): 1823-1830. Villabona-Arenas, C. J. and De Andrade Zanotto, P. M. (2011). Evolutionary history of Dengue virus type 4: Insights into genotype phylodynamics. Infection, Genetics and Evolution, 11 (5): 878-885. Von Heijne, G. (1984). How signal sequences maintain cleavage specificity. Journal of molecular biology, 173 (2): 243-251. Wang, E., Ni, H., Xu, R., Barrett, A. D., Watowich, S. J., Gubler, D. J. and Weaver, S. C. (2000). Evolutionary relationships of endemic/epidemic and sylvatic dengue viruses. Journal of virology, 74 (7): 3227-3234.

203

Chapter 6 References

Wang, P., Sidney, J., Dow, C., Mothe, B., Sette, A. and Peters, B. (2008). A systematic assessment of MHC class II peptide binding predictions and evaluation of a consensus approach. PLoS Comput Biol, 4 (4): e1000048. Wang, Q.-Y., Bushell, S., Qing, M., Xu, H. Y., Bonavia, A., Nunes, S., Zhou, J., Poh, M. K., De Sessions, P. F. and Niyomrattanakit, P. (2011). Inhibition of dengue virus through suppression of host pyrimidine biosynthesis. Journal of virology, 85 (13): 6548-6556. Wang, R., Doolan, D. L., Le, T. P., Hedstrom, R. C., Coonan, K. M., Charoenvit, Y., Jones, T. R., Hobart, P., Margalith, M. and Ng, J. (1998). Induction of antigen-specific cytotoxic T lymphocytes in humans by a malaria DNA vaccine. Science, 282 (5388): 476-480. Wang, R., Epstein, J., Baraceros, F. M., Gorak, E. J., Charoenvit, Y., Carucci, D. J., Hedstrom, R. C., Rahardjo, N., Gay, T. and Hobart, P. (2001). Induction of CD4+ T cell-dependent CD8+ type 1 responses in humans by a malaria DNA vaccine. Proceedings of the National Academy of Sciences, 98 (19): 10817-10822. Wang, S.-H., Syu, W.-J. and Hu, S.-T. (2004). Identification of the homotypic interaction domain of the core protein of dengue virus type 2. Journal of general virology, 85 (8): 2307- 2314. Wang, W.-K., Chen, H.-L., Yang, C.-F., Hsieh, S.-C., Juan, C.-C., Chang, S.-M., Yu, C.-C., Lin, L.-H., Huang, J.-H. and King, C.-C. (2006). Slower rates of clearance of viral load and virus- containing immune complexes in patients with dengue hemorrhagic fever. Clinical infectious diseases, 43 (8): 1023-1030. Webster, D. and Hill, A. V. (2003). Progress with new malaria vaccines. Bulletin of the World Health Organization, 81 (12): 902-909. Wei, Y., Qin, C., Jiang, T., Li, X., Zhao, H., Liu, Z., Deng, Y., Liu, R., Chen, S. and Yu, M. (2009). Translational regulation by the 3′ untranslated region of the dengue type 2 virus genome. The American journal of tropical medicine and hygiene, 81 (5): 817-824. Welsh, R. M. and Rothman, A. L. (2003). Dengue immune response: low affinity, high febrility. Nature medicine, 9 (7): 820-822. Wengler, G. (1989). Cell-associated West Nile flavivirus is covered with E+ pre-M protein heterodimers which are destroyed and reorganized by proteolytic cleavage during virus release. Journal of virology, 63 (6): 2521-2526. Wengler, G. and Wengler, G. (1981). Terminal sequences of the genome and replicatioe-form RNA of the flavivirus West Nile virus: Absence of poly (A) and possible role in RNA replication. Virology, 113 (2): 544-555. Wengler, G. and Wengler, G. (1993). The NS 3 nonstructural protein of flaviviruses contains an RNA triphosphatase activity. Virology, 197 (1): 265-273. Wengler, G., Wengler, G. and Gross, H. J. (1978). Studies on virus-specific nucleic acids synthesized in vertebrate and mosquito cells infected with flaviviruses. Virology, 89 (2): 423-437. Whitby, K., Pierson, T. C., Geiss, B., Lane, K., Engle, M., Zhou, Y., Doms, R. W. and Diamond, M. S. (2005). Castanospermine, a potent inhibitor of dengue virus infection in vitro and in vivo. Journal of virology, 79 (14): 8698-8706. Whitehead, S. S., Blaney, J. E., Durbin, A. P. and Murphy, B. R. (2007). Prospects for a dengue virus vaccine. Nature Reviews Microbiology, 5 (7): 518-528. Whitehead, S. S., Falgout, B., Hanley, K. A., Blaney Jr, J. E., Markoff, L. and Murphy, B. R. (2003). A live, attenuated dengue virus type 1 vaccine candidate with a 30-nucleotide deletion in the 3′ untranslated region is highly attenuated and immunogenic in monkeys. Journal of virology, 77 (2): 1653-1657.

204

Chapter 6 References

Wisseman, C. L., Sweet, B. H., Rosenzweig, E. C. and Eylar, O. R. (1963). Attenuated living type 1 dengue vaccines. The American Journal of Tropical Medicine and Hygiene, 12 (4): 620- 623. Wittke, V., Robb, T., Thu, H., Nisalak, A., Nimmannitya, S., Kalayanrooj, S., Vaughn, D., Endy, T., Holmes, E. and Aaskov, J. (2002). Extinction and rapid emergence of strains of dengue 3 virus during an interepidemic period. Virology, 301 (1): 148-156. Wu, H.-C., Jung, M.-Y., Chiu, C.-Y., Chao, T.-T., Lai, S.-C., Jan, J.-T. and Shaio, M.-F. (2003). Identification of a dengue virus type 2 (DEN-2) serotype-specific B-cell epitope and detection of DEN-2-immunized animal serum samples using an epitope-based peptide antigen. Journal of general virology, 84 (10): 2771-2779. Wu, S.-F., Lee, C.-J., Liao, C.-L., Dwek, R. A., Zitzmann, N. and Lin, Y.-L. (2002). Antiviral effects of an iminosugar derivative on flavivirus infections. Journal of virology, 76 (8): 3596-3604. Wu, S.-J. L., Grouard-Vogel, G., Sun, W., Mascola, J. R., Brachtel, E., Putvatana, R., Louder, M. K., Filgueira, L., Marovich, M. A. and Wong, H. K. (2000). Human skin Langerhans cells are targets of dengue virus infection. Nature medicine, 6 (7): 816-820. Wu, X., Hong, H., Yue, J., Wu, Y., Li, X., Jiang, L., Li, L., Li, Q., Gao, G. and Yang, X. (2010). Inhibitory effect of small interfering RNA on dengue virus replication in mosquito cells. Virol J, 7 (270): 1-8. Xie, X., Gayen, S., Kang, C., Yuan, Z. and Shi, P.-Y. (2013). Membrane topology and function of dengue virus NS2A protein. Journal of virology, 87 (8): 4609-4622. Yang, C.-C., Hsieh, Y.-C., Lee, S.-J., Wu, S.-H., Liao, C.-L., Tsao, C.-H., Chao, Y.-S., Chern, J.-H., Wu, C.-P. and Yueh, A. (2011). Novel dengue virus-specific NS2B/NS3 protease inhibitor, BP2109, discovered by a high-throughput screening assay. Antimicrobial agents and chemotherapy, 55 (1): 229-238. Yang, X. and Yu, X. (2009). An introduction to epitope prediction methods and software. Reviews in medical virology, 19 (2): 77-96. Yap, T. L., Xu, T., Chen, Y.-L., Malet, H., Egloff, M.-P., Canard, B., Vasudevan, S. G. and Lescar, J. (2007). Crystal structure of the dengue virus RNA-dependent RNA polymerase catalytic domain at 1.85-angstrom resolution. Journal of virology, 81 (9): 4753-4765. Yasser, E. and Honavar, V. (2010). Recent advances in B-cell epitope prediction methods. Immunome Res, 6 (Suppl 2): S2. Yin, Z., Chen, Y.-L., Schul, W., Wang, Q.-Y., Gu, F., Duraiswamy, J., Kondreddi, R. R., Niyomrattanakit, P., Lakshminarayana, S. B. and Goh, A. (2009). An adenosine nucleoside inhibitor of dengue virus. Proceedings of the National Academy of Sciences, 106 (48): 20435-20439. Yin, Z., Patel, S. J., Wang, W.-L., Wang, G., Chan, W.-L., Rao, K. R., Alam, J., Jeyaraj, D. A., Ngew, X. and Patel, V. (2006). Peptide inhibitors of Dengue virus NS3 protease. Part 1: Warhead. Bioorganic & medicinal chemistry letters, 16 (1): 36-39. Yon, C., Teramoto, T., Mueller, N., Phelan, J., Ganesh, V. K., Murthy, K. H. and Padmanabhan, R. (2005). Modulation of the nucleoside triphosphatase/RNA helicase and 5′-RNA triphosphatase activities of Dengue virus type 2 nonstructural protein 3 (NS3) by interaction with NS5, the RNA-dependent RNA polymerase. Journal of Biological Chemistry, 280 (29): 27412-27419. Young, A. C., Valadon, P., Casadevall, A., Scharff, M. D. and Sacchettini, J. C. (1997). The three- dimensional structures of a polysaccharide binding antibody to Cryptococcus neoformans and its complex with a peptide from a phage display library: implications for the identification of peptide mimotopes. Journal of molecular biology, 274 (4): 622-634.

205

Chapter 6 References

Zhang, H., Kolb, F. A., Jaskiewicz, L., Westhof, E. and Filipowicz, W. (2004a). Single processing center models for human Dicer and bacterial RNase III. Cell, 118 (1): 57-68. Zhang, Y., Zhang, W., Ogata, S., Clements, D., Strauss, J. H., Baker, T. S., Kuhn, R. J. and Rossmann, M. G. (2004b). Conformational changes of the flavivirus E glycoprotein. Structure, 12 (9): 1607-1618. Zhou, Y., Ray, D., Zhao, Y., Dong, H., Ren, S., Li, Z., Guo, Y., Bernard, K. A., Shi, P.-Y. and Li, H. (2007). Structure and function of flavivirus NS5 methyltransferase. Journal of virology, 81 (8): 3891-3903. Zinkernagel, R. M. (2004). On immunity against infections and vaccines: credo 2004. Scandinavian journal of immunology, 60 (1‐2): 9-13. Zybert, I. A., Van Der Ende-Metselaar, H., Wilschut, J. and Smit, J. M. (2008). Functional importance of dengue virus maturation: infectious properties of immature virions. Journal of General Virology, 89 (12): 3047-3051.

206

Research Papers

Papers Accepted/Published

 UmmarRaheel and Najam Us SaharSadafZaidi (2014). Inhibition of dengue virus 3 in

mammalian cell culture by synthetic small interfering RNAs targeting highly conserved

sequences. Tropical J Pharmaceutical Research. 13(10): 1621-1627. (IF:1)

 UmmarRaheel, Muhsin Jamal and NajamusSaharSadafZaidi (2015). A molecular

approach designed to limit the replication of mature DENV2 in host cells. Accepted in

Viral Immunology(IF:1.7)

 UmmarRaheel, Muhammad Faheem, Muhammad NasirRiaz, NaghmanaKanwal,

FarakhJaved, NajamusSaharSadafZaidi, IshtiaqQadri.Dengue fever in

IndianSubcontinent an overview. J Infect DevCtries. 2011 Apr 26;5(4):239-47. (IF: 1.2)

 UmmarRaheel, Muhammad Faheem, Muhammad NasirRiaz,

HashaamAkhtarSadafZaidi, IshtiaqQadri.Dengue virus: Advances in therapeutic

approaches and vaccine development”World Scientific Journal” Dengue Edition. (IF:2.7)

 Muhammad Faheem,UmmarRaheel, Muhammad NasirRiaz, NaghmanaKanwal,

FarakhJaved, NajamusSaharSadafZaidi, IshtiaqQadri. A Molecular Evaluation ofDengue

Virus Pathogenesis and Its Latest Vaccine Strategies. MolBiol Rep. 2011

Aug;38(6):3731-40. Epub 2010 Nov 24.(IF:2.1)

207