22
Accepted Manuscript Role of Intracellular Events in the Pathogenesis of Dengue; an Overview Bhawana Jain, Umesh. C. Chaturvedi, Amita Jain PII: S0882-4010(14)00037-0 DOI: 10.1016/j.micpath.2014.03.004 Reference: YMPAT 1473 To appear in: Microbial Pathogenesis Received Date: 27 December 2013 Revised Date: 17 March 2014 Accepted Date: 20 March 2014 Please cite this article as: Jain B, Chaturvedi UC, Jain A, Role of Intracellular Events in the Pathogenesis of Dengue; an Overview, Microbial Pathogenesis (2014), doi: 10.1016/ j.micpath.2014.03.004. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Role of intracellular events in the pathogenesis of dengue; An overview

  • Upload
    amita

  • View
    212

  • Download
    0

Embed Size (px)

Citation preview

Accepted Manuscript

Role of Intracellular Events in the Pathogenesis of Dengue; an Overview

Bhawana Jain, Umesh. C. Chaturvedi, Amita Jain

PII: S0882-4010(14)00037-0

DOI: 10.1016/j.micpath.2014.03.004

Reference: YMPAT 1473

To appear in: Microbial Pathogenesis

Received Date: 27 December 2013

Revised Date: 17 March 2014

Accepted Date: 20 March 2014

Please cite this article as: Jain B, Chaturvedi UC, Jain A, Role of Intracellular Events inthe Pathogenesis of Dengue; an Overview, Microbial Pathogenesis (2014), doi: 10.1016/j.micpath.2014.03.004.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Title: Role of Intracellular Events in the Pathogenesis of Dengue; an Overview 1

AUTHORS: Bhawana Jain*, Umesh. C. Chaturvedi*# and Amita Jain* 2

*Department of Microbiology, K.G. Medical University, Lucknow, India 3

# Indian Council of Medical Research, DHR, New Delhi, India 4

5

Address for correspondence: Dr Amita Jain, Professor, Department of Microbiology, K.G. 6

Medical University, Lucknow, India 7

e-mail: 8

Bhawana Jain – [email protected] (91 9450391235) 9

Umesh. C. Chaturvedi - [email protected] (91 9450913506) 10

Amita Jain – [email protected] (91 9415023928) 11

12

Word Count of Main text (including legend to figure, excluding tables, figure and 13

references): 4,305 14

Word Count of Abstract: 155 15

No. of Tables: 3 16

No. of figures: 1 17

No. of references: 99 18

-----------------------------------------------------------------------------------------------------. 19

20

21

22

#UCC is a Scientific Consultant of Indian Council of Medical Research, New Delhi 23

24

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Abstract 25

Dengue is one of the most important mosquito-borne viral diseases that are relentlessly 26

spreading in newer areas in the tropical and subtropical regions of the World. In last fifty 27

years, in spite of intensive and extensive investigations, pathogenesis of dengue is still not 28

clearly understood. Recently, the research focus is on studying the role of intracellular events 29

in pathogenesis of viral infections. Entry of virion in the host cell is followed by quick 30

succession of events, unfolded protein response, lipid bodies and lipophagy, endoplasmic 31

reticulum stress and recent demonstration of autophagy. The turbulence caused by these 32

events may result in clearance of the virus/ enhanced replication and survival of the host cell/ 33

apoptosis. Both, increased virus load and apoptosis of host cell may have pathological effects 34

on the host. In the present review, we have summed up the role of various intracellular events 35

in viral infections with special emphasis on Dengue virus infection. 36

37

38

Key Words: Dengue/ Autophagy/ Stress Granules/ Processing Bodies/ Lipid droplets 39

40

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

1. Introduction 41

Dengue virus (DENV) affects human of any age group, worldwide, including India1. 42

According to a recent estimate, there are around 390 million (95% credible interval 284–528) 43

dengue infections per year, of which 96 million (95% credible interval 67–136) manifest 44

apparently2. Dengue viruses occur as four antigenically related but distinct serotypes, 45

transmitted to humans by Aedes aegypti mosquitoes. These viruses generally cause either a 46

benign syndrome; dengue fever (DF), or a severe capillary leakage syndrome; dengue 47

hemorrhagic fever/ dengue shock syndrome (DHF/DSS)3. According to WHO 2009 48

classification severe Dengue has been differentiated into three subcategories; severe vascular 49

leakage, severe bleeding, and severe organ dysfunction4. The cardinal feature of DHF/DSS is 50

increased vascular permeability without morphological damage to capillary endothelium. 51

This results in extensive plasma leakage in various serous cavities of the host including 52

pleural, pericardial and peritoneal cavities and tissue spaces in patients with DHF, who may 53

go into profound shock (DSS)3,5. A number of mechanisms of pathogenesis of DHF/ DSS 54

have been discussed including, enhancing antibodies6,7, T cell-mediated response8 including 55

regulatory T cells9, various soluble mediators including a unique Cytotoxic Factor10,11 and 56

cytokines8,12,13, immune complex disease, antibodies cross-reacting with vascular 57

endothelium3, complement and its products, selection of virulent strains, viral virulence and 58

role of host genetics14. Among these the most plausible ones are the enhancing antibodies, 59

shift from Th1 to Th2 cell response and the memory T cells in a secondary infection, 60

resulting in a cytokine tsunami5. Extensive research has been done for more than fifty years 61

in area of dengue pathogenesis; still, the precise mechanism of DHF/DSS is not well 62

understood. In the last ten years or so, the research focus has shifted to intracellular events 63

during viral infections including DENV that may be translated to understand the pathogenesis 64

of severe disease. This review presents an overview of the role of intracellular events during 65

viral infection with special reference to dengue infection. 66

67

2. DENV replication and intracellular events: 68

Biologically diverse cell types, starting from insect cells to highly evolved mammalian cells 69

like endothelium and hepatocytes can be infected by DENV. The first interaction of DENV 70

with its host cell occurs via several putative receptors. They play an important role in 71

capturing, concentrating and transmitting infectious virions, leading to a cascade of events 72

that trigger the virus-cell membrane fusion15,16. A low pH-triggered conformational change of 73

Envelope (E) protein in endosomes leads to virus entry into the host cell through endocytosis 74

and uncoating. Capsid is released into the cell cytoplasm, where it dissociates and release 75

viral genome. Genome is a single RNA molecule of positive polarity and contains single 76

open reading frame (ORF), which is translated into a single large polyprotein. Polyprotein is 77

targeted to the endoplasmic reticulum (ER), where it is processed by virus and host encoded 78

proteases to form three structural proteins (Capsid protein C, a precursor for the membrane 79

protein PreM and envelope protein E) and seven non-structural proteins (NS1, NS2a, NS2b, 80

NS3, NS4a, NS4b, NS5), which help in replication, polyprotein processing and virion 81

assembly17. The replication takes place in virus induced vesicular membrane structures 82

associated with ER. Capsid protein covers the copies of genomes. Immature virions having 83

preM and E proteins on the surface bud into ER lumen18 and then transported through the 84

trans-Golgi network where cleavage of pre/M occurs making the virions infectious and 85

mature and is transported out of the cell by exocytosis19. 86

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

DENV exists as four heterologous serotypes termed serotypes 1, 2, 3 and 4 (DENV 1-4). 87

People once infected with one serotype of dengue virus are usually protected lifelong from 88

subsequent infection with the same serotype (homotypic infection)6. DHF occurs mostly in 89

persons infected with a second DENV serotype after an initial “primary” DENV infection 90

with a different serotype. The antibodies from the previous infection bind to the virus and 91

enhance its uptake by certain Fc receptor bearing monocytes/macrophages cells resulting in 92

high levels of viraemia. This phenomenon is known as antibody dependent enhancement 93

(ADE)6,7,20. Secondary infections cause forty times more DHF cases than primary infections. 94

There are many mechanisms suggested for DHF/DSS but nothing elaborates the pathogenesis 95

precisely. Recent studies have reported important findings of intimate interaction between 96

DENV and the host cell, which can be helpful in elucidating pathogenesis, like; a) autophagy; 97

b) lipid droplets and Lipophagy; c) unfolded protein response; d) Stress Granules and 98

Processing bodies. Further, strategies used by the virus to resist innate antiviral responses 99

have been discovered. Various mechanisms with reference to DENV infection will be 100

reviewed here. 101

2.A Autophagy 102

Autophagy is an intracellular catabolic system which degrades cytoplasmic components 103

within lysosomes. It has a specific role in elimination of micro-organisms, clearance of 104

intracellular proteins and organelles, cell death, and antigen presentation etc.21. The process 105

of autophagy is initiated when cell is subjected to pathogen infection. The signaling pathway; 106

Phosphatidyl Inositol 3-kinase (PI3K) and Beclin 1, gets activated. Beclin 1 first gets 107

separated from Beclin 2 which is a known anti apoptotic as well as anti autophagic protein, 108

and then induces autophagy. Kovacs et al have shown that inhibition of autophagy via Beclin 109

1 gene deletion in T cells results into extensive apoptosis of these cells upon T cell Receptor 110

(TCR) stimulation22. Beclin 1-deficient animals do not mount autoreactive T-cell responses. 111

This pathway leads to the formation of autophagosome, which is a key organelle of 112

autophagy. Certain autophagy related proteins (Atg) help in autophagosome formation. 113

Autophagosomes are formed in response to a number of stimuli, including host- and 114

pathogen-derived molecules, including toll-like receptor ligands and cytokines. Autophagy 115

can itself regulate the production and secretion of cytokines23. Autophagosome fuses with 116

lysosomes where lysosomal hydrolase degrades the inner membrane of autophagosome and 117

contain cytoplasm derived materials, known as autolysosomes or autophagolysosomes24. 118

Bhattacharya and Eissa have reviewed the role of autophagy and autoimmunity and have 119

concluded that autophagy, could modulate the induction or exacerbation of autoimmune 120

processes25. 121

2.A.(1) Role of autophagy in viral infection 122

Autophagy has been studied in a large number of viruses26 (Table 1 and 2). Viruses adopt 123

different mechanisms to induce or inhibit autophagy. Autophagy induction takes place by 124

following mechanisms i) by increase in autophagic flux, ii) by binding to the surface of host 125

cells, iii) by other intracellular events e.g. Endoplasmic Reticulum (ER) stress and Unfolded 126

Protein Response (UPR) (Table 1). Autophagy is inhibited (Table 1) by interfering with 127

either autophagosome formation, maturation and degradation or by degrading autolysosomes. 128

129

130

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Table 1: Impact of Viral infection on Autophagy 131

Mechanisms Viruses Viral molecules responsible for autophagy

Host molecules affected

References

A. Autophagy induction

by increase in autophagic flux Sindbis virus

Hepatitis C virus

-

-

Increases LC3 II

Increases LC3 II

27

28

by binding to the surface of target cells

Human Herpesvirus 6

Adenovirus B and D

Measles Virus

HIV 1

Vesicular Stomatitis Virus

Gp H-L-Q complex

trimeric fiber knob domain

gp41 subunit of viral envelope protein

-

Gp VSV-G

Binds to CD46

Binds to CD46

Binds to CD46

Binds to CXCR4 (CD4+) T cells

binds to TLR 7

-

-

29

30

31

by Endoplasmic Reticulum (ER) stress and Unfolded Protein Response (UPR)

Hepatitis C virus

Dengue Virus

-

NS4A

Induces PERK, ATF6, IRE1

-

32

33

B. Autophagy inhibition

by interfering with autophagosome formation

Kaposi Sarcoma Herpes Virus Murine Gamma Herpes Virus 68

Herpes Simplex Virus 1

vFLIP

M11

ICP34.5

target Beclin-1

encode viral Bcl 2 homologue

encode viral Bcl 2 homologue

34

35

36

by interfering with autophagosome maturation or degradation

HIV 1

Influenza A virus

Nef

Matrix (M2)

interact with Beclin-1

interact with Beclin-1

37

38

by autolysosomal degradation

Coxsackie Virus B3

Polio Virus

-

-

-

-

39

40

LC3 II= microtubule-associated protein 1 light chain 3; TLR= Toll like receptor; PERK= Protein Kinase R like 132 eIF2α kinase; ATF6= Activating Transcription Factor 6; IRE 1= Inositol Requiring Enzyme 1; NS= 133 Nonstructural; NEMO= NF κ B essential modulator; IRGM= Immunity associated GTPase family M; CD= 134 cluster of differentiation 135

136

137

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPTTable 2: Impact of Autophagy on viral pathogenesis 138

Mechanisms Viruses Viral molecules responsible for autophagy

Host molecules affected References

A. Degradation of viruses

by stimulating Innate Immunity

Vesicular Stomatitis Virus

Coxsackie Virus B3

-

-

production of IFN α by TLR7

TLR3 stimulation

-

-

by SLRs (Sequestosome 1/p62-like receptors)

Sindbis Virus

Herpes Simplex Virus 1

Capsid

-

degraded by p62

inhibited by SMURF1, novel adaptor

27

41

by stimulating Adaptive Immune response

Herpes Simplex Virus 1

EBV

Influenza A virus

-

EBNA1 protein

Matrix (M2)

presentation to MHC-I molecule

presentation to MHC-II mol

activates MHC II and CD4+ T cells

42

43

44

B. Promotion of viral replication

by formation of membranous replication factories

Polio Virus

Hepatitis C virus

Rotavirus

2BC and 3A proteins

NS5A, NS5B, nascent viral RNA

NS4P protein

-

-

-

45

46

47

by production of energy Dengue Virus - ATP production by Lipophagy

48

by promoting viral assembly and release

Hepatitis B Virus

HIV 1

Picorna virus

HBsAg

Gag, matrix (MA) protein p17

-

effects envelopment of viral particles

-

promotes nonlytic release

49

37

40

by suppressing antiviral innate immunity

Measles Virus

Hepatitis C virus

HIV 1

Capsid protein

NS3

Nef

target IRGM

target IRGM

target IRGM

50

50

50

LC3 II= microtubule-associated protein 1 light chain 3; TLR= Toll like receptor; PERK= Protein Kinase R like 139 eIF2α kinase; ATF6= Activating Transcription Factor 6; IRE 1= Inositol Requiring Enzyme 1; NS= 140 Nonstructural; NEMO= NF κ B essential modulator; IRGM= Immunity associated GTPase family M; CD= 141 cluster of differentiation 142

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

143

Consequently autophagic induction/inhibition leads to either virus degradation or promotion 144

of viral replication. Autophagy degrades viruses by following mechanisms (Table 2); i) by 145

stimulating innate immunity through Toll Like Receptors (TLRs), ii) by Sequestosome 1/p62-146

like receptors (SLRs) which are autophagic adaptors defined as pattern recognition receptors 147

(PRRs) that can selectively target a variety of pathogens for autophagic degradation, iii) by 148

stimulating adaptive immune response through MHC-I and MHC-II molecules. 149

Viral replication can also be promoted by following mechanisms (Table 2); i) by forming 150

Double Membrane Vesicles (DMVs) which help in replication, ii) by producing energy 151

through the process of lipophagy (discussed in section 2.B) iii) by enhancing viral assembly 152

and release, iv) by suppressing antiviral innate immunity. Innate immunity can be suppressed 153

by delivering cytosolic pathogen-associated molecular patterns (PAMPs) into the proximity 154

of endosomal pattern recognition receptors (PRRs) and MHC loading compartments and also 155

by directly trapping and degrading virions. 156

2.A.(2) Autophagy during dengue virus (DENV) infection 157

Some of the members of family Flaviviridae including DENV use autophagy for enhancing 158

their replication. Apoptosis is induced in macrophages by DENV, which is the primary site 159

for DENV replication. However, infected hepatocytes and epithelial cells which are the 160

secondary target of DENV infection do not die51-53. DENV evades innate immunity by 161

promoting autophagy in liver and kidney cells54. DENV2 induces autophagy in vitro in 162

MDCK renal epithelial cells, hepatocyte cell lines and other cells as well. These findings 163

were confirmed by inhibiting autophagy by the drug, 3-methyladenine as well as by siRNA, 164

targeting autophagy gene expression, which led to inhibition of viral replication. DENV 165

induced autophagosomes co-localize with LAMP 1, a marker of lysosomal fusion54. Other 166

studies also reported co-localization of DENV NS1 protein with autophagosome, LAMP 1 167

and ribosomal protein L2855. M6P-R, one of the endosomal markers, co-localizes with 168

autophagosomes, indicating that autophagosome also fuse with endosomes forming 169

amphisome. Both, autophagosome and amphisome are double membrane vesicles (DMVs) 170

and DENV is known to replicate on virally induced DMVs. DENVs replication on 171

amphisomes possibly influences virus entry and replication. Inhibition of lysosomal fusion 172

with autophagosomes increases replication of DENV 255. NS 4A (Nonstructural protein 4A) 173

of DENV2 is the viral mediator of autophagy upregulation and protection against apoptosis33. 174

DENV utilizes PI3K dependent mechanism to upregulate autophagy. Proteins Beclin 1 and 175

Atg 5 are required for protection of virus infected cells55. Another study suggested that 176

autophagy stimulates DV replication by suppressing innate immune response of the host 177

through Unfolded Protein Response (UPR)/ autophagy pathway28. 178

Role of autophagy is also studied in DENV 3 infection. LAMP 1 co-localizes with 179

autophagosomes like DENV2 but lysosomal fusion inhibitor decreases DENV3 replication, 180

suggesting role of mature autophagosomes in DENV3 infection. Inhibition of formation of 181

autolysosomes by L-arginine, induces the accumulation of autophagosomes and amphisomes. 182

This mechanism stimulates DENV2 replication and inhibits DENV3 replication, suggesting 183

DENV2 uses amphisomes whereas DENV3 uses both amphisomes and autolysosomes for 184

viral RNA replication55,56. Welsch et al showed 3D structures of DENV2 replication 185

complex, which was actually the contiguous invagination of Endoplasmic reticulum (ER)57. 186

He further suggested indirect role of functional components of autophagy in DENV 187

replication, maturation and infectivity by inhibiting autophagy using spautin-1 (specific 188

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

and potent autophagy inhibitor-1), a novel inhibitor and stimulator of autophagy in DENV 189

infection58. Spautin-1 treatment generated heat sensitive and non infectious DENV particles 190

denoting major effect of autophagy on DENV cell cycle. 191

Monocytes are the critical cell in the dengue disease. They are rarely infected directly, but are 192

highly susceptible to antibody enhanced infection59. Panyasrivanit et al have reported that in 193

monocytic cells induction of autophagy reduced viral yield while inhibition of autophagy 194

only slightly increased levels of viruses and there are distinct cell type specific differences in 195

the DENV-autophagy interaction60. 196

There are several studies elucidating the role of autophagy in DENV replication, however, 197

the precise mechanism by which DENV induces autophagy is still unclear. More work is 198

required to further characterize the role(s) and contribution of autophagy to the various 199

aspects of DENV replication, in different cell types. 200

2.B Lipid Droplets and Lipophagy 201

Lipids are stored in a cell in the form of lipid droplets (LDs) and are metabolized by 202

cytoplasmic neutral hydrolases. Lipid droplets (LDs) are dynamic organelles containing 203

neutral lipid core. They arise from Endoplasmic Reticulum (ER) as a result of various stress 204

signals. Lipid metabolism occurring through the lysosomal degradative pathway of 205

autophagy, known as lipophagy, regulates intracellular lipid stores and cellular levels of free 206

lipids such as fatty acids and energy homeostasis. An important role of lipophagy is in the 207

maintenance of adequate levels of β-oxidation to supply ATP which may be responsible for 208

the regulation of cell resistance to death stimuli and in cellular trans-differentiation. Recently 209

LDs have received special attention as they have been found to be associated with viral 210

diseases (eg. HCV, HBV and Rotavirus). Studies on HCV, have shown the positive effect of 211

LDs and associated ER system in promoting viral replication61. Core protein of HCV has 212

been shown to be involved in LDs distribution in perinuclear region and helping viral 213

replication and assembly62-64. Study on DENV demonstrated that LDs sequestrated unfolded 214

proteins and prevented the accumulation of lipotoxic non-esterified fatty acid by diverting 215

them to cytosol for degradation65. Later on Capsid protein of DENV was observed to be 216

present over LDs. In early stages of infection, capsid protein sequestration takes place while 217

later on; LDs serve as platform for assembly of virus. Therefore LDs have indirect role on 218

DENV replication by regulating the capsid protein availability66. Another study also 219

demonstrated the role of ER rearrangement and expansion in early stages of DENV infection 220

which involved reabsorption of LDs67. 221

Lipophagy functions in a number of different cell types, and appears to be a common 222

pathway of cellular lipid metabolism68. Lipophagy eliminates intracellular pathogens, but 223

viruses may subvert the lipophagic pathway to promote their own survival. For example, 224

hepatitis C virus (HCV)69 and hepatitis B virus70 upregulate autophagy which in turn 225

promotes viral replication. The core protein of HCV is known to attach to LD for promoting 226

viral assembly through an unknown mechanism71. Studies with DENV have demonstrated 227

that autophagy increases viral replication through lipophagy. Further, upregulation of 228

autophagy promotes lipid breakdown for supply of energy for viral replication72. DENV 229

infection of hepatoma cells promote the formation of autophagosomes which associate with 230

LDs resulting in stimulation of lipophagy in infected cells. Reduction of lipid droplets as well 231

as cellular triglycerides upon DENV infection indicates that regulation of lipid metabolism by 232

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

autophagy is a major component facilitating DENV replication. Cellular triglycerides that are 233

stored in lipid droplets are depleted by DENV-induced autophagosomes, resulting into 234

increased β-oxidation and energy production. This showed that a virus can trigger autophagy 235

to modulate cellular physiology48,72. Heaton et al have identified three cellular pathways 236

needed for DENV replication: autophagy, actin polymerization, and fatty acid biosynthesis48. 237

Further, a key enzyme in this pathway, fatty acid synthase (FASN), is relocalized to sites of 238

DENV replication by DENV nonstructural protein 3 (NS3). Purified recombinant NS3 also 239

stimulates the activity of FASN in vitro. Thus, it is suggested that DENV co-opts the fatty 240

acid biosynthetic pathway to establish its replication complexes48. In conclusion, this novel 241

mechanism of lipophagy helps DENV not only to promote replication but also to evade 242

immune system of the host by channelizing autophagosomes towards lipid metabolism73. 243

However, despite our improved understanding of lipid metabolism and ER rearrangement, 244

more research is required to precisely delineate the steps leading to DENV replication 245

promotion by LDs or Lipophagy. This will help in development of new therapeutic 246

modalities against DENV. Inhibition of FASN or any of the enzymes/ proteins of autophagy 247

pathway can inhibit DENV replication73. 248

2.C Unfolded Protein Response (UPR) 249

The unfolded protein response (UPR) is cellular response to ER stress which causes 250

accumulation of unfolded or misfolded proteins in the lumen of the endoplasmic reticulum 251

(ER) in a stressed cell. In virus-infected cells, the cellular translation machinery is 252

orchestrated by the infecting virus to produce large amounts of viral proteins, which 253

ultimately perturbs ER homeostasis and causes ER stress74. UPR restores normal function of 254

the cell by attenuating protein translation and activates the signaling pathways to increase the 255

production of molecular chaperones required in protein folding. If these aims are not 256

achieved, the UPR leads to apoptosis. Several viruses are shown to induce UPR. 257

Herpesviruses induce ER stress and activate UPR signaling pathways which help in assembly 258

of infectious particles75. HCV induces the UPR, which activates autophagic pathway by 259

initiating autophagosome formation76. Recently, it has been discovered that autophagy plays 260

a regulatory role in controlling the innate immune response against intracellular pathogens77. 261

HCV induces autophagy through UPR activation. Autophagy is required for promoting HCV 262

RNA replication in human hepatoma cells. This is achieved by suppressing innate antiviral 263

immunity28 through upgradation of HCV-derived pathogen-associated molecular pattern 264

(PAMP), which mediates anti viral response. DENV, like HCV, also utilizes the ER or ER-265

derived membrane structure as the primary site of RNA replication and assembly. Thus, it is 266

conceivable that DENV infected cells experience ER stress and the UPR. Ke and Chen 267

(2011) in the same study showed suppression of DENV-PAMP RNA-induced IFN β 268

promoter activation by UPR inducers, which causes exploitation of UPR-autophagy pathway 269

to escape innate immune response in both DENV and HCV infections28. DENV infection of 270

A549 cells elicits an UPR at the level of translation attenuation and activation of specific 271

pathways. Specific serotypes of DENV can modulate the UPR with different selectivity. The 272

modulators of UPR such as Salubrinal inhibit DENV replication78. Pena and Harris have 273

reported a time-dependent activation of the UPR by DENV-2 which can override inhibition 274

of translation, prevent apoptosis, and prolong the viral life cycle79. Other studies have shown 275

that modulation of intracellular membrane architecture of the cell early after DENV-2 276

infection is driven by viral protein expression and does not require the induction of the UPR 277

and sterol-regulatory-element-binding-protein-2 (SREBP-2) pathways67. Studies on the 278

impact on UPR following direct or ADE infection of THP-1 cells with all the four serotypes 279

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

of DENV showed that the ADE infection with epidemic DENV-2 (high-UPR-gene-280

expressing strains) correlated with severe disease80. 281

2.D Stress Granules and Processing Bodies 282

Viral infection, along with other types of stresses, alters the cells’ protein expression system. 283

In order to adapt to the stress, the supressed expression of constitutive proteins lead to change 284

in translation pathway and diversion of mRNA from polysomes to cytoplasmic granules. 285

These granules are of various types in eukaryotic cells but Stress granules (SG) and 286

Processing bodies (PB) are of special interest in virus infection. 287

During cell stress, untranslated mRNA accumulates in discrete cytoplasmic foci called stress 288

granules (SGs) that have roles in inhibition and degradation of host mRNAs. SGs appear to 289

protect RNAs from harmful conditions, and may safe-guard the information coded in their 290

RNA sequence. These molecules can be stored, degraded, or re-initiate translation81. 291

Processing bodies (P-bodies, PB) are another type of granules packed with translationally 292

silenced mRNPs and enzymes of RNA decay machine. PBs may promote SG assembly82,83. 293

SGs and PBs are actively linked and are believed to transiently bind and exchange mRNP 294

along with remodeling of the mRNP protein constituents, playing roles in decay of unwanted 295

mRNA, nonsense mRNA, AU-rich element mediated mRNA and silencing of microRNA 296

induced mRNA. 297

2.D.(1) Role of SG and PB in viral replication 298

Virus infection interferes with a number of cell processes which directly induce stress 299

responses such as transient global inhibition of protein synthesis to enhance cell survival by 300

restricted consumption of nutrients and energy. Although SG formation is frequently induced 301

by virus infection, many differences exist in the dynamics and outcome of the stress 302

responses induced by various viruses84. Viruses have evolved measures to prevent 303

sequestration of viral mRNA into translationally silenced mRNPs that aggregate in SG and 304

get decayed in PBs. Virus families can be organized into groups on the basis of mechanisms 305

adopting to facilitate their replication by modulating SGs and PBs (Table 3). The gene 306

expression and resources to damage repair pathways may be redirected. The mechanism of 307

SG formation is not understood clearly but may involve remodeling of mRNP which 308

incorporates new proteins. These proteins nucleate SGs and affect mRNP transport on 309

microtubules. Quick reactivation of translation on recovery from stress may be facilitated by 310

SGs. SGs sequester components of apoptotic signal transduction pathways, therefore, may 311

help in cell survival during stress85. Intracellular stress occurs during viral infection which 312

reduces translation through activation of enzymes like eIF2 kinases and changes active 313

polysome mRNPs into stalled translation initiation complex mRNPs. Nucleation of several 314

stress granule marker proteins (viz. G3BP, Tia-1/TIAR, and HDAC6 plus) transport on 315

microtubules (MT) resulting into aggregation of translation initiation complex mRNPs in 316

stress granules, through a series of complex steps. Decapping and decay can occur within or 317

outside PBs. 318

319

320

321

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Table 3: Stress Granules (SGs) and Processing Bodies (PBs) studied in different viral 322

infections 323

Viruses Involved molecules and processes References 1. Inhibition of Stress Granules (SGs)

A. Activation of eIF2α kinase

Polio Virus - 86

Alpha Virus - - Orthoreo Virus - - CMV eIF4G mediated 87

Rotavirus eIF4E, S6, eIF2α phosphorylation 88

Influenza A Virus blocks eIF2α phosphorylation 89

Other Reovirideae eIF4G, eIF4E, eIF3

90,91

Chikungunya Virus - 92

B. Blocking loss of cellular SG formation in response to Oxidative stress

Polio virus Cleavage of SG nucleating protein G3BP, PABP

86,93

West Nile Virus Sequestration of SG nucleating protein TIA-1/TIAR

94

Dengue Virus Sequestration of SG nucleating protein TIA-1/TIAR, G3BP1, Caprin 1, USP 10

94,95

Herpes Simplex Virus 1

Sequestration of SG nucleating protein TIA-1/TIAR

96

HTLV 1 Sequestration of SG nucleation and maintenance protein HDAC 6

97

HIV 1 Sequestration of PABP 1,Staufen 1 98

Rotavirus Sequestration of TIA-1 88

Other Reovirideae Sequestration of TIA-1, TIAR, G3BP 90,91

2. Disruption of Processing Bodies (PBs) Hepatitis C virus Binds with DDX 3, RCK/p 54, Lsm

1, Pat L1

99

Polio Virus - 86,93

Coxsackie Virus - - West Nile Virus - -

3. Alteration of Stress Granule (SG) response Respiratory Syncytial Virus

eIF2α phosphorylation by PKR 100

Vaccinia Virus subvert SG-nucleating proteins 101

324

G3BP= Ras-Gap-SH3 domainbinding protein; TIA-1= T cell intracellular antigen- ; TIAR= T cell 325

intracellular antigen-1 related protein; eIF2α = eukaryotic translation initiation factor 2α; PKR= 326

protein kinase R 327

328

2.D.(2) SGs and PBs in dengue virus (DENV) infection 329

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

DENV is a single stranded positive sense RNA of ≈ 11 kb containing single ORF and is the 330

only viral mRNA produced during viral replication. DENV RNA replication complexes are 331

shown to locate in the perinuclear region of infected cells. Researchers have observed co-332

localization of SG nucleating protein i.e. T cell intracellular antigen-1 (TIA-1) and related 333

protein (TIAR) with DV NS3 and dsRNA in perinuclear region only94. The kinetics of TIAR 334

accumulation was found to be similar to genomic RNA synthesis. DENV components 335

interact with TIAR, facilitate viral RNA synthesis and inhibit SG formation. This prevents 336

stallation of host translation machinery. Co localization of many proteins related to SGs 337

(G3BP 1, G3BP 2, Caprin 1 and USP 10) and PBs (DDX 6 also known as RCK/p 54) with 338

DENV serotype 2 viral components is known95. These proteins bind to DENV at 3’UTR 339

region at different sites, but 3’UTR region of DENV 2 is an important site for PB and SG 340

assembly and DDX 6 had been demonstrated as a pro-viral protein helping in assembly or 341

release of infectious particles. 342

Such studies demarcate the definitive role of SG and PB in DV infection and it is not anti 343

viral, since it is enhancing DV survival. However, further studies may delineate the exact 344

mechanism and help the investigators to find some robust tool to control dengue by 345

modulating these bodies. 346

3. Summary of Intracellular Events and DHF/DSS pathogenesis 347

A number of mechanisms have been considered to explain the pathogenesis of DHF/DSS. 348

The most favored ones are increased virus load by replication via enhancing antibodies, virus 349

virulence, shift from Th1 to Th2 cell response and the memory T cells in a secondary 350

infection, resulting in cytokine tsunami producing severe dengue disease. However, 351

everything is not explained by these mechanisms, therefore a fascinating hypothesis emerged, 352

which stated that intracellular events and pathways may have significant role in DENV 353

infection; mild or severe. The intracellular event that follows entry of DENV in the host cells 354

may indicate the outcome in the form of recovery or severe disease. Viruses are obligate 355

intracellular parasites. Their survival and expansion depends on the health of the host cells 356

providing necessary supporting factors to the viruses. In virus-infected cells, the cellular 357

translation machinery is hijacked by the infecting virus to produce large amounts of viral 358

proteins. The accumulation of unfolded or misfolded proteins, leads to highly specific 359

signaling pathways i.e. the UPR to restore normal ER functions. If this overload is not 360

resolved, the UPR induces ER stress-associated programmed cell death, apoptosis. 361

Autophagy, LD formation and Lipophagy are also among these defensive mechanisms of 362

host against viral infection. To combat such defense system of host, certain viruses like 363

DENV, modulate such events in its favor and use such system to enhance its replication or 364

survival, as an adaptation trick. NS4A protein of DENV induces autophagy by UPR. The 365

UPR activates autophagic pathway which promotes virus replication in cells. This increased 366

viral replication is favored by formation of membranous replication factories also known as 367

double membrane vesicles (DMV) or by production of energy. DMVs provide platform for 368

viral replication. In DENV infection, lipid metabolism leading to the production of ATP by β 369

oxidation (Lipophagy) promotes viral replication. Innate immunity of the host is also 370

tempered by these events to help DENV infection (Figure 1). 371

372

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT 373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

Fig 1: Role of various intracellular pathways in pathogenesis of dengue virus infection 403

404

405

DV Infection

Autophagy upregulation ER stress

Autophagosome formation

UPR

Phosphorylation

eIF2α

Stalled ternary complex

mRNA translation Inhibition

LD

β Oxidation of lipid

ATP production

Increased DV replication SG

PB

Active

polysome

PERK

IRE1

ATF6

PERK

PKR

GCN2

HR1

Inhibition of DV replication

Assembly restricted

Increased DV survival

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Legend to Figure 1: 406

407

In response to stress caused by DV infection, Endoplasmic reticulum (ER) of host cells, 408

which is otherwise responsible for the synthesis, maturation and proper folding of a wide 409

range of proteins, leads to the induction of inter-organelle signaling pathways. These 410

pathways regulate translation and gene expression through an elaborate adaptive response 411

known as the unfolded protein response (UPR). The UPR functions through the PKR-like ER 412

kinase (PERK), kinase/ endoribonuclease inositol requiring 1 (IRE1)/X-box binding protein 1 413

(XBP1), and/or activation transcription factor 6 (ATF6) pathways which together inhibit 414

translation. PERK along with PKR (Protein Kinase R), GCN2 and HRI lead to 415

phosphorylation of eukaryotic translation initiation factor (eIF) 2α (eIF2α) and arrest of 416

translation due to stalled ternary complex (2(eIF2)+GTPNmet-tRNAi met). The 417

nontranslated mRNA along with proteins is organized into specialized RNA granules, Stress 418

Granules (SGs) and Processing Bodies (PBs). DV sequesters SG proteins thus restricting SG 419

assembly and therefore enhancement in DV survival into host cells occur. Another pathway 420

used by DV is Autophagy which involves sequestration of intact organelles (e.g., 421

mitochondria) and portions of the cytosol via a membrane referred to as the phagophore 422

(derived from ER). This phagophore expands to form double-membrane vesicles, termed 423

autophagosomes. Subsequently, autophagosomes mature by fusing with endosomes and/or 424

lysosomes to form autolysosomes, where degradation of the internal contents occurs by 425

resident lysosomal hydrolases. DV induces degradation of LDs via autophagy in order 426

tomobilize triglycerides that are used for energy (ATP) production via β-oxidation. 427

428

DV: Dengue virus; LD: Lipid Droplets; UPR: Unfolded protein response; ER: Endoplasmic 429

reticulum; SG: Stress Granules; PB: Processing Bodies 430

431

432

4. Conclusions 433

The pathogenesis of dengue is very complex. Impact of intracellular events including 434

autophagy, stress granules and processing bodies, unfolded protein response etc. is not yet 435

fully elucidated in the pathogenesis of dengue. Impact of Dengue virus infection on various 436

intracellular processes is highly complex too and needs to be explained in detail. Delineation 437

of interaction of DENV with each mechanism upto the molecular level is highly needed to 438

find the suitable targets for novel and highly promising anti viral therapeutic modalities, 439

being introduced each day in the scientific field. 440

Acknowledgements 441

Authors acknowledge financial support from ICMR Grant no. 5/8/7/14/2009-ECD-I phase-II 442

and ICMR Grant no. 5/8/7/17/2010-ECD-I. UCC is Scientific Consultant of ICMR/DHR, 443

New Delhi. 444

Conflict of interest: None 445

446

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

References: 447

448

1. Gupta N, Srivastava S, Jain A, Chaturvedi UC (2012) Dengue in India. Indian J Med 449

Res136(3):373-90 450

2. Bhatt S, Gething PW, Brady OJ et al (2013) The global distribution and burden of 451

dengue. Nature, DOI:10.1038/nature12060 452

3. Basu A, Chaturvedi UC (2008) Vascular endothelium: the battlefield of dengue 453

viruses. FEMS Immunol. Med. Microbiol53:287–299. 454

4. World Health Organization, 2009. Dengue: Guideline for Diagnosis, Treatment, 455

Prevention and Control. Geneva: World Health Organization 456

5. Jain A, Chaturvedi UC (2010) Dengue in infants: an overview. FEMS Immunol Med 457

Microbiol59(2):119-30. 458

6. Halstead SB (2003) Neutralization and antibody-dependent enhancement of dengue 459

viruses. Adv Virus Res60:421-467 460

7. Halstead SB (2009) Antibodies determine virulence in dengue. Ann N Y Acad Sci 461

1171 Suppl 1:E48-56. 462

8. Chaturvedi UC, Raghupathy R, Pacsa AS et al (1999) Shift from a Th1-Type 463

Response to Th2-Type in Dengue Haemorrhagic Fever Curr Sci76; 63-69. 464

9. Chaturvedi UC, Shrivastava S, Tripathi RK, Nagar R (2007) Dengue virus-specific 465

suppressor T cell: Current perspectives. FEMS Immunol Med50: 285-299. 466

10. Mukerjee R, Chaturvedi UC, Dhawan R (1995) Dengue Virus-induced human 467

cytotoxic factor: production by peripheral blood leucocytes in vitro. Clin Exp 468

Immunol101:261-267. 469

11. Mukerjee R, Chaturvedi UC, Vaughn DW, Kalayanarooj S (1997) Purification and 470

pathogenicity of the cytotoxic factor from the cases of dengue haemorrhagic fever. 471

Curr Sci72:494-501. 472

12. Chaturvedi UC, Agarwal R, Elbishbishi EA, Mustafa AS (2000) Cytokine Cascade 473

in Dengue Haemorrhagic Fever: Implications for Pathogenesis. FEMS Immunol Med 474

Microbiol28:183-188 475

13. Rathakrishnan A, Wang SM, Hu Y, Khan AM, Ponnampalavanar S, Lum 476

LC, Manikam R, Sekaran SD (2012) Cytokine expression profile of dengue patients at 477

different phases of illness. PLoS One7(12):e52215. doi: 478

10.1371/journal.pone.0052215. 479

14. Chaturvedi UC, Nagar R, Shrivastava S (2006a) Dengue and dengue haemorrhagic 480

fever: implications of host genetics. FEMS Immunol Med Microbiol47:155-166. 481

15. Basu A, Chaturvedi UC (2010) Recent advances in understanding the intracellular 482

responses to dengue virus infection (Invited editorial) Future Virol5:255–257. 483

16. Chaturvedi UC, Nagar R, Shrivastava S (2006b) Macrophage and dengue virus: friend 484

or foe? Indian J Med Res124:23-40 485

17. Bartenachlager R, Miller S. Molecular aspects of Dengue virus replication. Future 486

Microbiol. 2008 Apr;3:155-165 487

18. Mukhopadhyay S, Kuhn RJ, Rossmann MG. A structural perspective of the flavivirus 488

life cycle. Nat. Rev. Microbiol. 2005Jan;3(1):13-22 489

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

19. Mackenzie JM, Jones MK, Westaway EG. Markers for trans Golgi membranes and 490

the intermediate compartment localize to induced membranes with distinct replication 491

functions in flavivirus-infected cells. J. Virol. 1999 Nov;73(11):9555-9567 492

20. Chaturvedi UC, Shrivastava, R, Nagar, R.(2005) Dengue Vaccines: Prospects and 493

Problems. Indian J Med. Res121:639-52 494

21. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y (2004) In vivo 495

analysis of autophagy in responose to nutrient starvation using transgenic mice 496

expressing a fluorescent autophagosome marker. Mol Biol Cell15:1101–1111. 497

22. Kovacs JR, Li C, Yang Q, Li G, Garcia IG, Ju S, Roodman DG, Windle JJ, Zhang. X, 498

Lu B. (2012) Autophagy promotes T-cell survival through degradation of proteins of 499

the cell death machinery. Cell Death Differ. 19:144-152. 500

23. Harris J. (2011). Autophagy and cytokines. Cytokine. ;56:140-144. 501

24. Mizushima, N. Autophagy: Process and function. Genes. Dev. 2007, 21, 2861-2873. 502

25. Bhattacharya A, Eissa NT. (2013). Autophagy and autoimmunity crosstalks. Front. 503

Immunol. 15;4:88. doi: 10.3389/fimmu.2013.00088. 504

26. Chiramel AI, Brady NR, Bartenschlager R (2013) Divergent Roles of Autophagy in 505

Virus Infection. Cells2:83-104; doi:10.3390/cells2010083 506

27. Orvedahl A, MacPherson S, Sumpter R Jr, Talloczy Z, Zou Z, Levine B (2010) 507

Autophagy protects against Sindbis virus infection of the central nervous system. Cell 508

Host Microbe7:115-127. 509

28. Ke PY, Chen SS (2011) Activation of the unfolded protein response and autophagy 510

after hepatitis C virus infection suppresses innate antiviral immunity in vitro. J Clin 511

Invest121:37-56. 512

29. Joubert PE, Meiffren G, Gregoire IP et al (2009) Autophagy induction by the 513

pathogen receptor CD46. Cell Host Microbe6: 354-366. 514

30. Denizot M, Varbanov M, Espert L, Robert-Hebmann V, Sagnier S, Garcia E, Curriu 515

M, Mamoun R.and Blanco J, Biard-Piechaczyk, M (2008) HIV-1 gp41 fusogenic 516

function triggers autophagy in uninfected cells. Autophagy 008, 4, 998-1008. 517

31. Nakamoto M, Moy RH, Xu J, Bambina S, Yasunaga A, Shelly SS, Gold B, Cherry S 518

(2012) Virus recognition by Toll-7 activates antiviral autophagy in Drosophila. 519

Immunity36:658-667. 520

32. Kroemer G, Marino G, Levine, B (2010) Autophagy and the integrated stress 521

response. Mol Cell40: 280-293. 522

33. McLean JE, Wudzinska A, Datan E, Quaglino D, Zakeri Z (2011) Flavivirus NS4A-523

induced autophagy protects cells against death and enhances virus replication. J Biol 524

Chem286:22147-22159. 525

34. Liang CEX, Jung JU (2008) Downregulation of autophagy by herpesvirus Bcl-2 526

homologs. Autophagy4: 268-272. 527

35. Ku B, Woo JS, Liang C, Lee KH, Hong HS, E X, Kim KS, Jung JU, Oh BH (2008) 528

Structural and biochemical bases for the inhibition of autophagy and apoptosis by 529

viral BCL-2 of murine gamma-herpesvirus 68. PLoS Pathog 4, e25. 530

36. Orvedahl A, Alexander D, Talloczy Z, Sun Q, Wei Y, Zhang W, Burns D, Leib DA, 531

Levine B (2007) HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 532

autophagy protein. Cell Host Microbe1: 23-35. 533

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

37. Kyei GB, Dinkins C, Davis AS et al (2009) Autophagy pathway intersects with HIV-534

1 biosynthesis and regulates viral yields in macrophages. J Cell Biol186: 255-268. 535

38. Gannage M, Dormann D, Albrecht R et al (2009) Matrix protein 2 of influenza A 536

virus blocks autophagosome fusion with lysosomes. Cell HostMicrobe6: 367-380. 537

39. Kemball CC, Alirezaei M, Flynn CT, Wood MR, Harkins S, Kiosses WB, Whitton JL 538

(2010) Coxsackievirus infection induces autophagy-like vesicles and 539

megaphagosomes in pancreatic acinar cells in vivo. JVI84: 12110-12124. 540

40. Jackson WT, Giddings TH, Jr, Taylor MP, Mulinyawe S, Rabinovitch M, Kopito RR, 541

Kirkegaard K (2005) Subversion of cellular autophagosomal machinery by RNA 542

viruses. Plos Biol3: e156. 543

41. Orvedahl A, Sumpter R Jr, Xiao G et al (2011) Image-based genome-wide siRNA 544

screen identifies selective autophagy factors. Nature480: 113-117. 545

42. English L, Chemali M, Duron J, Rondeau C, Laplante A, Gingras D, Alexander D, 546

Leib D, Norbury C, Lippe R, Desjardins M (2009) Autophagy enhances the 547

presentation of endogenous viral antigens on MHC class I molecules during HSV-1 548

infection. Nat Immunol10: 480-487. 549

43. Paludan C, Schmid D, Landthaler M, Vockerodt M, Kube D, Tuschl T, Munz C 550

(2005) Endogenous MHC class II processing of a viral nuclear antigen after 551

autophagy. Science307: 593-596. 552

44. Schmid D, Pypaert M, Munz C (2007) Antigen-loading compartments for major 553

histocompatibility complex class II molecules continuously receive input from 554

autophagosomes. Immunity26:79-92. 555

45. Schlegel A, Giddings TH Jr, Ladinsky MS, Kirkegaard K (1996). Cellular origin and 556

ultrastructure of membranes induced during poliovirus infection. JVI70: 6576-6588. 557

46. Sir D, Kuo CF, Tian Y, Liu HM, Huang EJ, Jung JU, Machida K, Ou JH (2012) 558

Replication of hepatitis C virus RNA on autophagosomal membranes. J Biol 559

Chem287: 18036-18043. 560

47. Berkova Z, Crawford SE, Trugnan G, Yoshimori T, Morris, AP, Estes MK (2006). 561

Rotavirus NSP4 induces a novelvesicular compartment regulated by calcium and 562

associated with viroplasms. JVI, 80, 6061-6071. 563

48. Heaton NS, Randall G (2011) Dengue virus and autophagy. Viruses3(8):1332-341. 564

49. Li J, Liu Y, Wang Z, Liu K, Wang Y, Liu J, Ding H, Yuan Z (2011) Subversion of 565

cellular autophagy machinery by hepatitis B virus for viral envelopment. JVI 85: 566

6319-6333. 567

50. Gregoire IP, Richetta C, Meyniel-Schicklin L et al (2011) IRGM is a common target 568

of RNA viruses that subvert the autophagy network. PLoS Pathog7: e1002422. 569

51. Tonry J. H., Xiao S. Y., Siirin M., Chen H., da Rosa A. P., Tesh R. B. (2005) Am. J. 570

Trop. Med. Hyg. 72, 320–324. [PubMed: 15772329] 571

52. Siirin M. T., Duan T., Lei H., Guzman H., da Rosa A. P., Watts D. M., Xiao S. Y., 572

Tesh R. B. (2007) Am. J. Trop. Med. Hyg. 76, 299–306. [PubMed: 17297039] 573

53. Murray K., Walker C., Herrington E., Lewis J. A., McCormick J., Beasley D. W., 574

Tesh R. B., Fisher-Hoch S. (2010) J. Infect. Dis. 201, 2–4.[PMCID: 575

PMC2791189] [PubMed: 19961306] 576

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

54. Lee YR, Lei HY, Liu MT, Wang JR, Chen SH, Jiang-Shieh YF, Lin YS, Yeh TM, Liu 577

CC, Liu HS (2008) Autophagic machinery activated by dengue virus enhances virus 578

replication. Virology374:240–248 579

55. Panyasrivanit M, Khakpoor A, Wikan N, Smith DR (2009) Co-localization of 580

constituents of the dengue virus translation and replication machinery with 581

amphisomes. J Gen Virol90:448–456 582

56. Khakpoor A, Panyasrivanit M, Wikan N, Smith DR (2009) A role for 583

autophagolysosomes in dengue virus 3 production in HepG2 cells. J GenVirol90: 584

1093–1103 585

57. Welsch S, Miller S, Romero-Brey I, Merz A, Bleck CK, Walther P, Fuller SD, 586

Antony C, Krijnse-Locker J, Bartenschlager R (2009) Composition and three-587

dimensional architecture of the dengue virus replication and assembly sites. Cell Host 588

Microbe5:365-375. 589

58. Mateo R, Nagamine CM, Spagnolo J, Méndez E, Rahe M, Gale M Jr, Yuan 590

J, Kirkegaard K (2013) Inhibition of cellular autophagy deranges dengue virion 591

maturation. J Virol87(3):1312-21 592

59. Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM (2010) Intrinsic 593

antibody-dependent enhancement of microbial infection in macrophages: disease 594

regulation by immune complexes. Lancet Infect Dis10(10):712-22 595

60. Panyasrivanit M, Greenwood MP, Murphy D, Isidoro C, Auewarakul P, Smith DR 596

(2011) Induced autophagy reduces virus output in dengue infected monocytic 597

cells. Virology418:74–84. 598

61. Roingeard P, Depla M (2011) The birth and life of lipid droplets: learning from the 599

hepatitis C virus. Biol Cell 103: 223–231. 600

62. Roingeard P, Hourioux C, Blanchard E, Prensier G (2008) Hepatitis C virus budding 601

at lipid droplet-associated ER membrane visualized by 3D electron microscopy. 602

Histochem Cell Biol 130: 561–566. 603

63. Boulant S, Douglas MW, Moody L, Budkowska A, Targett-Adams P, et al. (2008) 604

Hepatitis C virus core protein induces lipid droplet redistribution in a microtubule- 605

and dynein-dependent manner. Traffic 9: 1268–1282. 606

64. Miyanari Y, Atsuzawa K, Usuda N, Watashi K, Hishiki T, et al. (2007) The lipid 607

droplet is an important organelle for hepatitis C virus production. Nat Cell Biol 9: 608

1089–1097. 609

65. Hapala I, Marza E, Ferreira T (2011) Is fat so bad? Modulation of endoplasmic 610

reticulum stress by lipid droplet formation. Biol Cell 103: 271–285. 611

66. Samsa MM, Mondotte JA, Iglesias NG, Assuncao-Miranda I, Barbosa-Lima G, Da 612

Poian AT et al (2009) Dengue virus capsid protein usurps lipid droplets for viral 613

particle formation. PLoS Pathog5: e1000632. 614

67. Peña J, Harris E (2012) Early dengue virus protein synthesis induces extensive 615

rearrangement of the endoplasmic reticulum independent of the UPR and SREBP-2 616

pathway. PLoS One(6):e38202. doi: 10.1371/journal.pone.0038202. 617

68. Liu K, Czaja MJ (2013) Regulation of lipid stores and metabolism by lipophagy. Cell 618

Death and Differentiation20: 3–11. 619

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

69. Ait-Goughoulte M, Kanda T, Meyer K, Ryerse JS, Ray RB, Ray R (2008) Hepatitis C 620

virus genotype 1a growth and induction of autophagy. J Virol82: 2241–2249. 621

70. Sir D, Tian Y, Chen WL, Ann DK, Yen TS, Ou JH (2010) The early autophagic 622

pathway is activated by hepatitis B virus and required for viral DNA replication. Proc 623

Natl Acad Sci USA107:4383–4388. 624

71. McLauchlan J (2009) Hepatitis C virus: viral proteins on the move. Biochem Soc 625

Trans 37: 986–990. 626

72. Heaton NS, Randall G (2010) Dengue virus-induced autophagy regulates lipid 627

metabolism. Cell Host Microbe8: 422–432. 628

73. Heaton NS, Perera R, Berger KL, Khadka S, Lacount DJ, Kuhn RJ, Randall G (2010) 629

Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral 630

replication and increases cellular fatty acid synthesis. Proc Natl Acad Sci 631

USA107:17345–17350 632

74. Zhang L, Wang A (2012) Virus-induced ER stress and the unfolded protein 633

response. Front Plant Sci3:293. doi: 10.3389/fpls.2012.00293. 634

75. Lee DY, Lee J, Sugden B. The unfolded protein response and autophagy: 635

herpesviruses rule! J Virol. 2009;83(3):1168–1172. 636

76. Schroder M. Endoplasmic reticulum stress responses. Cell Mol Life Sci. 637

2008;65(6):862–894. 638

77. Munz C. Enhancing immunity through autophagy. Annu Rev Immunol. 2009;27:423–639

449. 640

78. Umareddy I, Pluquet O, Wang QY, Vasudevan SG, Chevet E, Gu F (2007) Dengue 641

virus serotype infection specifies the activation of the unfolded protein response. 642

Virol J4:91. 643

79. Peña J, Harris E (2011) Dengue virus modulates the unfolded protein response in a 644

time-dependent manner. J Biol Chem286(16):14226-36. 645

80. Paradkar PN, Ooi EE, Hanson BJ, Gubler DJ, Vasudevan SG (2011) Unfolded protein 646

response (UPR) gene expression during antibody-dependent enhanced infection of 647

cultured monocytes correlates with dengue disease severity. Biosci Rep31(3):221-30. 648

81. Anderson P, Kedersha N (2009) RNA granules: post-transcriptional and epigenetic 649

modulators of gene expression. Nat Rev Mol Cell Biol10: 430–436 650

82. Eulalio A, Behm-Ansmant I, Izaurralde E (2007) P bodies: at the crossroads of post-651

transcriptional pathways. Nat Rev Mol Cell Biol8:9–22 652

83. Buchan JR, Muhlrad D, Parker R (2008) P bodies promote stress granule assembly in 653

Saccharomyces cerevisiae. J Cell Biol183: 441–455 654

84. Lloyd RE (2012) How Do Viruses Interact with Stress-Associated RNA Granules? 655

PLoS Pathog8(6): e1002741. doi:10.1371/journal.ppat.1002741 656

85. White JP, Lloyd RE (2012) Regulation of stress granules in virus systems. Trends 657

Microbiol20:175–183 658

86. Piotrowska J, Hansen SJ, Park N, Jamka K, Sarnow P, et al. (2010) Stable Formation 659

of compositionally unique stress granules in virus-infected cells. J Virol 84: 3654–660

3665 661

87. Isler JA, Skalet AH, Alwine JC (2005) Human Cytomegalovirus Infection Activates 662

and Regulates the Unfolded Protein Response. J Virol79:6890-6899. 663

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

88. Montero H et al. (2008) Rotavirus infection induces the phosphorylation of eIF2alpha 664

but prevents the formation of stress granules. J Virol82:1496–1504 665

89. Khaperskyy DA, Hatchette TF, McCormick C (2011) Influenza A virus inhibits 666

cytoplasmic stress granule formation. Faseb J DOI: 10.1096/fj.11-196915 667

90. Smith JA, Schmechel SC, Raghavan A et al. (2006) Reovirus induces and benefits 668

from an integrated cellular stress response. J Virol80: 2019–2033 669

91. Qin Q, Hastings C, Miller CL (2009) Mammalian orthoreovirus particles induce and 670

are recruited into stress granules at early times postinfection. J Virol 83:11090–11101. 671

92. Rathore AP, Ng ML, Vasudevan SG (2013) Differential unfolded protein response 672

during Chikungunya and Sindbis virus infection: CHIKV nsP4 suppresses eIF2α 673

phosphorylation. Virol J10:36 674

93. White JP, Cardenas AM, Marissen WE, Lloyd RE (2007) Inhibition of cytoplasmic 675

mRNA stress granule formation by a viral proteinase. Cell Host Microbe 2: 295–305. 676

94. Emara MM, Brinton MA (2007) Interaction of TIA-1/TIAR with West Nile and 677

dengue virus products in infected cells interferes with stress granule formation and 678

processing body assembly. Proc Natl Acad Sci USA104(21):9041–9046. 679

95. Ward A M, Bidet K, Yinglin A, Ler S G, Hogue K, Blackstock W, Gunaratne J,. 680

Garcia-Blanco M A (2011) Quantitative mass spectrometry of DENV-2 RNA-681

interacting proteins reveals that the DEAD-box RNA helicase DDX6 binds the DB1 682

and DB2 3' UTR structures. RNA Biology8:1173-1186. 683

96. Esclatine A et al. (2004) The UL41 protein of herpes simplex virus mediates selective 684

stabilization or degradation of cellular mRNAs. Proc Natl Acad Sci U.S.A. 101: 685

18165–18170 686

97. Legros S et al. (2011) The HTLV-1 Tax protein inhibits formation of stress granules 687

by interacting with histone deacetylase 6. Oncogene 30:4050–4062 688

98. Abrahamyan LG, Chatel-Chaix L, Ajamian L et al. (2010) Novel Staufen1 689

ribonucleoproteins prevent formation of stress granules but favour encapsidation of 690

HIV-1 genomic RNA. J Cell Sci123:369–383 691

99. Ariumi Y, Kuroki M, Kushima Y, Osugi K, Hijikata M, et al. (2011) Hepatitis C virus 692

hijacks P-body and stress granule components around lipid droplets. J Virol 85: 6882–693

6892. 694

100. Lindquist ME et al. (2011) Activation of protein kinase R is required for 695

induction of stress granules by respiratory syncytial virus but dispensable for viral 696

replication. Virology413: 103–110 697

101. Simpson-Holley M, Kedersha N, Dower K, Rubins KH, Anderson P, et al. 698

(2011) Formation of antiviral cytoplasmic granules during orthopoxvirus infection. J 699

Virol 85: 1581–1593 700

701

702

703

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

• Role of intracellular events is very important in dengue virus (DENV) infection

• Endoplasmic reticulum (ER) is the key point to provide double membrane structures

• DENV exploits autophagy to increase its replication

• Assembly of Stress granules is decreased by DENV to increase its survival in host cells

• Unfolded protein response (UPR) also plays a critical role