AVR 3649
No. of Pages 10, Model 5G
16 June 2015 Antiviral Research xxx (2015) xxx–xxx 1
Contents lists available at ScienceDirect
Antiviral Research journal homepage: www.elsevier.com/locate/antiviral
2
Review
5 4 6
Cellular and molecular mechanisms of chikungunya pathogenesis
7 8 9 10 11 12 1 2 4 6 15 16 17 18 19 20 21 22 23 24 25
Fok-Moon Lum a,b, Lisa F.P. Ng a,b,c,⇑ a
Singapore Immunology Network, Agency for Science, Technology and Research (A⁄STAR), Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore c Institute of Infection and Global Health, University of Liverpool, United Kingdom b
a r t i c l e
i n f o
a b s t r a c t
Article history: Received 6 May 2015 Revised 27 May 2015 Accepted 16 June 2015 Available online xxxx
Chikungunya virus (CHIKV) is an arthropod-borne virus that causes chikungunya fever, a disease characterized by the onset of fever and rashes, with arthralgia as its hallmark symptom. CHIKV has re-emerged over the past decade, causing numerous outbreaks around the world. Since late 2013, CHIKV has reached the shores of the Americas, causing more than a million cases of infection. Despite concentrated efforts to understand the pathogenesis of the disease, further outbreaks remain a threat. This review highlights important findings regarding CHIKV-associated immunopathogenesis and offers important insights into future directions. This article forms part of a symposium in Antiviral Research on ‘‘Chikungunya discovers the New World.’’ Ó 2015 Published by Elsevier B.V.
Keywords: Chikungunya virus Immune response Immunopathogenesis Pathology
27 28 29 30 31 32 33 34 35 36 37
38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57
Contents 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16.
Introduction . . . . . . . . . . . . . The virus . . . . . . . . . . . . . . . . Replication cycle . . . . . . . . . Route of infection. . . . . . . . . Target cells . . . . . . . . . . . . . . Apoptosis and autophagy . . Importance of Type I IFN . . . Cytokines and chemokines . IL-6 and osteoclastogenesis . Monocytes/macrophages . . . NK cells . . . . . . . . . . . . . . . . . Dendritic cells. . . . . . . . . . . . T cells . . . . . . . . . . . . . . . . . . B cells . . . . . . . . . . . . . . . . . . Future perspectives . . . . . . . Uncited references . . . . . . . . Acknowledgements . . . . . . . References . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . .
00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00
58 59
60
1. Introduction
61
Chikungunya virus (CHIKV) is an alphavirus belonging to the Togaviridae family that was first isolated from a human patient in
62
⇑ Corresponding author at: Singapore Immunology Network, Agency for Science, Technology and Research (A⁄STAR), Singapore. E-mail address: lisa_ng@immunol.a-star.edu.sg (L.F.P. Ng).
Tanzania in 1952. It is transmitted mainly by the Aedes agypti and Aedes albopictus mosquitoes. Infection causes a self-limiting febrile illness known as chikungunya fever (CHIKF) with symptoms such as myalgia, fever and rashes. Patients also typically exhibit polyarthralgia, which is a hallmark of the disease. Symptoms usually appear after an incubation period of 4–7 days. While many of the symptoms disappear within the following week, arthralgia may persist in some patients for up to a few years (Her et al., 2009; Kam
http://dx.doi.org/10.1016/j.antiviral.2015.06.009 0166-3542/Ó 2015 Published by Elsevier B.V.
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
63 64 65 66 67 68 69 70
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 2
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
83
et al., 2009). In some cases, CHIKF has been associated with neurological complications such as myeloradiculopathy and meningoencephalitis (Borgherini et al., 2007; Chandak et al., 2009). However, direct neurovirulence and neuroinvasiveness remain to be investigated. In this article, we discuss interactions between CHIKV and the host immune response, with a focus on the balance between protection and pathology in defining CHIKV pathogenesis. Our paper forms part of a symposium in Antiviral Research on ‘‘Chikungunya discovers the New World.’’ Readers interested in a general review of the disease should refer to the article ‘‘Chikungunya: Evolutionary history and recent epidemic spread’’ (Weaver and Forrester, 2015) published in this symposium.
84
2. The virus
85
CHIKV is an enveloped virus that is approximately 70 nm in diameter in neutral pH and contains a 11.8 kb single-stranded, positive-sense RNA genome (Strauss and Strauss, 1994; Leung et al., 2011; Rashad et al., 2014). The genome consists of a 50 methylated terminal cap untranslated region (UTR), followed by RNA coding for 4 non-structural proteins (nsP1–4) and 5 structural
71 72 73 74 75 76 77 78 79 80 81 82
86 87 88 89 90
proteins (C–E3–E2–6K–E1), and a 30 terminal poly-A tail (Strauss and Strauss, 1994; Leung et al., 2011; Teng et al., 2011). The non-structural proteins and structural proteins, governed by 2 separate open-reading frames (ORFs), contribute to the propagation of new virions (Schwartz and Albert, 2010; Leung et al., 2011; Rashad et al., 2014). The non-structural genes encode for nsP1 and nsP3, helicase (nsP2) and polymerase (nsP4). These proteins associate to form the viral replication complex needed for downstream viral genome replication (Solignat et al., 2009; Schwartz and Albert, 2010). On the other hand, the structural capsid protein is involved in the formation of the icosahedral fenestrated nucleocapsid of a mature virion, in which the viral RNA genome will be contained (Solignat et al., 2009; Schwartz and Albert, 2010; Rashad et al., 2014). The E1 and E2 glycoproteins associate as a heterodimer before being incorporated onto the surface of the mature virion as trimeric spikes and are involved in the attachment and entry of the virion into susceptible target cells during subsequent infection. A total of 80 trimeric spikes are present on the surface of a mature virion (Voss et al., 2010). The role of the 6K protein remains ambiguous, but it is thought to be involved in virus assembly and budding (Leung et al., 2011;
Fig. 1. CHIKV replication cycle. The virus enters susceptible cells through endocytosis, mediated by an unknown receptor. As the endosome is acidic, conformational changes occur resulting in the fusion of the viral and host cell membranes, causing the release of the nucleocapsid into the cytoplasm, The RNA genome is first translated into the 4 nsPs, which together will form the replication complex and assist in several downstream processes (depicted by dashed arrowed line). Subsequently, the genome is replicated to its negative-sense strand, which in turn will be used as a template for the synthesis of the 49S viral RNA and 26S subgenomic mRNA. The 26S subgenomic mRNA will be translated to give the structural proteins (C–pE2–6K–E1). After a round of processing by serine proteases, the capsid is released into the cytoplasm. The remaining structural proteins are further modified post-translationally in the endoplasmic reticulum and subsequently in the Golgi apparatus. E1 and E2 associate as a dimer and are transported to the host plasma membrane, where they will ultimately be incorporated onto the virion surface as trimeric spikes. Capsid protein will form the icosahedral nucleocapsid that will contain the replicated 49S genomic RNA before being assembled into a mature virion ready for budding. During budding the virions will acquire a membrane bilayer from part of the host cell membrane.
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx 113 114 115 116 117 118 119 120 121
Teng et al., 2011). In addition, it has also been reported that alphavirus 6K protein is part of a cation-selective ion channel (Melton et al., 2002), which has the potential to affect membrane permeability. Finally, E3 protein is important in directing the structural proteins to the endoplasmic reticulum (ER) for proper assembly and spike formation via its cognate interactions with E2 glycoprotein (Snyder and Mukhopadhyay, 2012). It has also been reported that alphavirus E3 protein could play a role in pH protection during virus formation and budding (Uchime et al., 2013).
122
3. Replication cycle
123
Upon entry into target cells (Fig. 1), conformational changes in the virus envelope take place, due to the acidic endosomal environment. With these conformational changes, the E1 glycoprotein will be exposed and this mediates fusion of the CHIKV membrane with the host endosomal membrane, with the nucleocapsid being released into the cytoplasm in the process. The free viral RNA then gets translated into a polyprotein that gets cleaved into the nsPs 1– 4. These nsPs associate to form a functional viral replication complex, which will then go on to generate the full-length negative sense RNA intermediate. This RNA intermediate will be used as a template for downstream synthesis of the 49S genomic RNA as well as the 26S subgenomic mRNA.
124 125 126 127 128 129 130 131 132 133 134
3
The 26S subgenomic mRNA is translated to give the Capsid–p E2–6K–E1 polyprotein that is further processed by serine proteases to release the capsid into the cytoplasm. The remaining proteins are then directed to the ER in which they will undergo further post-translational modifications. pE2 will then be cleaved in the Golgi to give E2 and E3 glycoproteins. E1 and E2 glycoproteins will heterodimerize and get transported to the host cell plasma membrane where they would be incorporated onto the virion surface as trimeric spikes. Capsid protein associates in the cytoplasm, forming the icosahedral nucleocapsid that contains the 49S genomic RNA. Finally, mature virions are assembled at the plasma membrane, and bud out of the infected host cell, during which the mature virions will acquire a membrane bilayer from the host cell plasma membrane (Strauss and Strauss, 1994; Leung et al., 2011; Rashad et al., 2014). Typically, CHIKV replication occurs between 8 to 16 h post-infection (Teng et al., 2011), while in vertebrate cells, virions are typically produced less than 8 h after infection at 37 °C (Sourisseau et al., 2007).
135
4. Route of infection
154
CHIKV infects the host through inoculation at the skin via bites from infected mosquitoes. It can infect skin resident cells such as
155
Fig. 2. Pathogenesis of CHIKV infection: viral dissemination and impact on host immune response. (1) CHIKV infection occurs through the bite of an infected mosquito, resulting in a (2) dermal infection phase. Skin resident cells become infected, allowing for the initial rounds of replication. (3) Direct inoculation of virus into the circulation can also occur via mosquito bites. (4) Subsequently, the virus disseminates to draining lymph nodes, where it further replicates before being (5) released into the circulation. (6) It then disseminate to peripheral organs, such as the spleen and muscles, where further replication takes place. The figure is adapted from (Ong et al., 2014) with permission.
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153
156
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 4 157 158 159 160 161 162 163 164 165 166 167 168
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
the skin fibroblasts and dermal macrophages (Sourisseau et al., 2007; Couderc et al., 2008; Kam et al., 2009). This will lead to the first round of virus replication and trigger an initial host immune response to contain the virus at the skin (Kupper and Fuhlbrigge, 2004; Schilte et al., 2010). Despite the immune response, the virus rapidly disseminates into the lymph node before further dissemination to other tissues (e.g., muscles) via the circulatory system (Kam et al., 2009). Virus replication occurs at these peripheral tissues, resulting in the viremic phase of the disease (Fig. 2). During this phase, virus can be easily transmitted to mosquitoes via a blood meal. Viremia is normally cleared within 2–10 days postinfection (Panning et al., 2008; Kam et al., 2009).
169
5. Target cells
170
The identification of susceptible cell types is important for understanding the pathogenesis of CHIKV infection. Studies of cellular tropism have reported that the virus can infect and replicate in a variety of cell lines and adherent cells, such as endothelial, epithelial and fibroblastic primary cells (Sourisseau et al., 2007; Salvador et al., 2009; Wikan et al., 2012). It was also demonstrated that CHIKV was capable of infecting human muscle satellite cells, but not in the differentiated myotubes (Ozden et al., 2007). On the other hand, various groups have reported contrasting results regarding the infectability of peripheral blood mononuclear cells (PBMCs) (Sourisseau et al., 2007; Her et al., 2010; Teng et al., 2012). Animal models have been developed to aid in the investigation of CHIKV infection. Adult C57BL/6 wildtype animals inoculated at the ventral side of the footpad recapitulated the rheumatic manifestation commonly observed in infected humans (Gardner et al., 2010; Lum et al., 2013; Teo et al., 2013). In these animals, virus was also identified in the spleen, lymph nodes, liver and muscles, complementing observations obtained in in vitro studies. The profile of systemic viremia and blood count reflected that of an infected patient (Gardner et al., 2010; Lum et al., 2013; Teo et al., 2013). Studies in non-human primates have also reported the presence of fever, rash and pronounced joint pain during the acute phase of the disease, with the peak of viremia at 1–2 days after infection (Chen et al., 2010; Labadie et al., 2010; Messaoudi et al., 2013). These symptoms were accompanied by transient acute lymphopenia and neutropenia. Virus was detected in numerous organs and tissues such as the skin, brain, liver, muscle, lymph nodes, spleen and joint-connective tissues. Persistent infection was also detected in the joints, muscles, and splenic macrophages, as well as in endothelial cells lining the liver sinusoids (Chen et al., 2010; Labadie et al., 2010; Messaoudi et al., 2013). However, it remains to be further investigated if this observation is a true reflection of an infected patient, although there has been a report of CHIKV persistence in the perivascular synovial macrophages of a chronically infected patient some 18 months after illness onset (Hoarau et al., 2010). The effect of age on CHIKV persistence should also be investigated as it has been shown that the virus persists in elderly rhesus macaques due to a defective antiviral immune response (Messaoudi et al., 2013). Moreover, given the increased episodes of neurological complications and the presence of viral antigen in the cerebrospinal fluid of patients (Chandak et al., 2009; Economopoulou et al., 2009; Kashyap et al., 2010), further development of animal models would serve as valuable research tools to investigate neurotropism and neuroinvasiveness. In fact, it has been shown using neonatal mice and mice with an abrogated Type I Interferon (IFN) signaling pathway, that the virus can disseminate into the central nervous system, targeting the choroid plexuses and leptomeninges (Couderc et al., 2008).
171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219
6. Apoptosis and autophagy
220
Apoptosis has been demonstrated to increase the spread of virus from apoptotic infected cells to neighboring uninfected cells (Krejbich-Trotot et al., 2011a). The virus has also been shown to hide in apoptotic blebs as an evasion strategy, escaping cellular host responses such as extracellular antibodies (Long and Heise, 2015). More recently, autophagy (Glick et al., 2010) was reported to be induced during active infection, further increasing viral replication (Krejbich-Trotot et al., 2011b). However, while autophagy was shown to increase CHIKV replication in a human cell line (Krejbich-Trotot et al., 2011b), a decrease in replication was observed in mouse embryonic fibroblasts (Joubert et al., 2012). This species-specific difference was later demonstrated to be due to interaction of the human autophagy receptor NDP52 with the viral nsP2 to promote viral replication, which was not observed with the mouse orthologue of NDP52 (Judith et al., 2013). The cross-talk between autophagy and apoptosis to mediate CHIKV protection and pathogenesis remains to be further established.
221
7. Importance of Type I IFN
239
The first line of host defense against pathogen infection is the innate immune response. Given that the adaptive immune response requires over a week to develop, the innate immune response would thus be crucial in defending against CHIKV infection. The type I IFN pathway is a vital innate anti-viral pathway, and it has been well-reported that CHIKV infection elicits the type I IFN response alongside the production of other pro-inflammatory cytokines (Gardner et al., 2010; Labadie et al., 2010; Chow et al., 2011; Wauquier et al., 2011; Teng et al., 2012). Particularly, IFNa and IFNb have been reported to play crucial anti-viral roles in both CHIKV infected humans and mice, as well as in a series of cell lines derived from both species (Sourisseau et al., 2007; Couderc et al., 2008; Gardner et al., 2010; Schilte et al., 2012; Teng et al., 2012). The importance of Type I IFN response was further demonstrated where CHIKV infection was lethal in mice deficient in Type I IFN receptors (Couderc et al., 2008). Further experiments in chimeric mice demonstrated that Type I IFN produced by non-hematopoietic cells is vital in the effective clearance of CHIKV (Schilte et al., 2010). It has also been revealed that the nsP2 protein of CHIKV has the ability to shut off Type I IFN signaling leading to a decreased expression of anti-viral mediators (Fros et al., 2010). Induction of Type I IFN is triggered through pattern recognition receptors (PRRs) following the recognition of structurally conserved pathogen-associated molecular patterns (PAMPs) (Medzhitov and Janeway, 2000; Janeway and Medzhitov, 2002). Cellular PRRs such as the Toll-like receptors (TLRs) and cytosolic retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), such as the melanoma differentiation-associated protein 5 (MDA5) and RIG-I have been reported to be play crucial roles in mediating Type I IFN production during CHIKV infection (Stetson and Medzhitov, 2006; Schilte et al., 2010; Teng et al., 2011; Her et al., 2015). Upon infection and replication, double-stranded RNA (dsRNA) intermediates picked up by both MDA5 and RIG-I will kick-start a downstream signaling cascade which further involves the CARD adaptor inducing IFNb adaptor protein (CARDIF) and crucial interferon-regulatory factors (IRF) 3 and 7 (Kawai et al., 2005; Honda et al., 2006; Stetson and Medzhitov, 2006). The activation of this pathway ultimately leads to the production of Type I IFNs, which act either as a paracrine or autocrine signal to further amplify the anti-viral signaling cascade mediated through Janus
240
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238
241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308
kinases (JAK) and signal transducers and activators of transcription (STAT) (Decker et al., 2002; Stetson and Medzhitov, 2006). This is followed by the production of interferon-stimulated genes (ISG), such as Viperin, GADD44, Tetherin (BST-2), ISG15, Mx proteins, protein kinase R (PKR) and 20 ,50 -oligoadenylate synthase 3 (OAS3), which have reported anti-CHIKV properties (Bréhin et al., 2009; Schoggins and Rice, 2011; Schoggins et al., 2011; Werneke et al., 2011; White et al., 2011; Clavarino et al., 2012; Teng et al., 2012; Jones et al., 2013). Anti-viral ISGs can also be induced through TLR signaling (Kawai and Akira, 2010). The importance of TLR3 in hematopoietic cells in controlling CHIKV infection has recently been revealed (Her et al., 2015). Given that TLR3 recognizes dsRNA, it was shown in the absence of TLR3 that CHIKV-infected mice suffered a more pronounced disease accompanied with higher viremia. This complemented an earlier report that the loss of TRIF (Toll/IL-1 resistance domain-containing adaptor inducing IFNb), an essential adaptor molecular for TLR3 signaling (Kawai and Akira, 2010), induced a more severe disease outcome in mice (Rudd et al., 2012). In PBMCs obtained from clinically CHIKV-infected patients, it was reported that high levels of IFNa, IFNb and TLR3 transcripts were detected when compared to non-infected controls (Teng et al., 2012; Her et al., 2015). These observations further substantiate the importance of these pathways in controlling CHIKV progression, which is likely to be controlled through the combined effects of two or more sensor pathways, deriving from both hematopoietic and non-hematopoietic cells (Schilte et al., 2010; Her et al., 2015).
309
8. Cytokines and chemokines
310
330
Extensive studies have been performed to characterize the immune response of CHIKV-infected patient cohorts. In these studies, pro-inflammatory cytokines such as IL-6, monocyte chemotactic protein-1 (MCP-1) and IFNc were found to be elevated during the acute phase of the disease in several patient cohorts (Ng et al., 2009; Hoarau et al., 2010; Chow et al., 2011; Kelvin et al., 2011; Wauquier et al., 2011). Positive correlation was also observed between the expression of IL-6 or MCP-1 and the high viral load in CHIKV-infected patients (Chow et al., 2011). Interestingly, IL-6 and GM-CSF were also observed to associate with persistent arthralgia (Hoarau et al., 2010; Chow et al., 2011). More recently, a meta-analysis comparative study demonstrated that pro-inflammatory cytokines such as IFNa, IFNc, IL-2, IL-2R, IL-6, IL-7, IL-12, IL-15, IL-17 and IL-18; anti-inflammatory cytokines such as IL-1Ra, IL-4 and IL-10; chemokines: granulocyte colony-stimulating factor (GM-CSF), IP-10, MCP-1, monokine induced by gamma interferon (MIG), macrophage inflammatory protein (MIP) 1a and MIP-1b; and growth factor: basic fibroblast growth factor (FGF) formed a generic acute CHIKV signature in all the different patient cohorts around the world (Teng et al., 2015).
331
9. IL-6 and osteoclastogenesis
332
The exact role of IL-6 in CHIKV infection has yet to be fully elucidated. However, it has been shown using primary osteoblasts prepared from the knee-joint of a healthy male subject that these cells could be infected by the virus, and upon infection produced high levels of IL-6 and RANKL that were maintained up to 40 days post-infection (Noret et al., 2012). This was further supported by the detection of IL-6 in the infected joint of a chronically ill patient, further indicating that IL-6 could drive persistent arthralgia (Hoarau et al., 2010). Given that the IL-6 receptor is expressed on osteoblasts, IL-6 may act in an autocrine manner to induce more of its own production in a positive feedback manner (Li et al.,
311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329
333 334 335 336 337 338 339 340 341 342
5
2008). It was also shown that there was a repression of osteoprotegerin (OPG) production in infected osteoblasts (Noret et al., 2012). The high level of RANKL coupled with a decreased presence of OPG activates osteoclasts and drives osteoclastogenesis (Hofbauer et al., 1999), a mechanism that has been proposed to be responsible for CHIKV-induced rheumatic symptoms (Noret et al., 2012). Using a closely related alphavirus, the Ross River Virus (RRV), it was demonstrated that RRV infection in primary human osteoblasts led to the production of high levels of pro-inflammatory cytokines, including IL-6 and MCP-1. This was further coupled with a disrupted RANKL/OPG ratio in the synovium of RRV patients. The importance of IL-6 in driving bone loss was then determined in a RRV mouse model, where neutralization of IL-6 prevented abnormality of the RANKL/OPG ratio (Chen et al., 2014).
343
10. Monocytes/macrophages
358
IL-6 is also known to strongly induce the expression of MCP-1 (Romano et al., 1997), a monocyte/macrophage chemo-attractant (Lu et al., 1998). As shown by numerous studies, MCP-1 is strongly associated with the acute phase of CHIKV infection both in infected humans and animals (Chen et al., 2010, 2015; Gardner et al., 2010; Hoarau et al., 2010; Teng et al., 2015). In CHIKV-infected animals, this elevated level of MCP-1 was accompanied by increased infiltration of monocytes into the site of inflammation (Gardner et al., 2010; Labadie et al., 2010; Poo et al., 2014a). The importance of monocytes/macrophages in driving CHIKV-induced pathology was further illustrated in mice treated with liposomes containing clodronate, which depletes macrophages (Gardner et al., 2010). In these treated mice, CHIKV-associated disease pathology was ameliorated, which suggests the critical role of macrophages in driving CHIKV pathology. However, it should be noted that the effect of clodronate is not specific to macrophages as it is known to deplete all phagocytic cells including dendritic cells and neutrophils (Zhang et al., 2002). MCP-2 and MCP-3 were also reported to be present in high amounts in the joints of CHIKV-infected animals (Chen et al., 2015). Treatment with Bindarit, a MCP inhibitor (Bhatia et al., 2005) was demonstrated to completely abolish CHIKV-induced pathology (Rulli et al., 2011; Chen et al., 2015). This is further complemented by studies in the related arthritogenic RRV, where monocytes/macrophages were shown to be the main drivers of pathology (Lidbury et al., 2000, 2008; Herrero et al., 2011, 2013; Taylor et al., 2013). Monocytes/macrophages have also been suggested to be the cellular vehicle for virus dissemination as well as being a cellular reservoir for persistent CHIKV infection (Her et al., 2010; Hoarau et al., 2010; Labadie et al., 2010). CHIKV infection in CCR2 / animals resulted in a more severe, prolonged and erosive arthritis, with no effect on viral replication (Poo et al., 2014a). Loss of CCR2, which is the receptor for MCP-1, caused a drastic change in the profile of infiltrating immune cells, coupled with a dysregulation of both pro- and anti-inflammatory pathways.
359
11. NK cells
395
Alongside the increase of monocyte/macrophage infiltration into the inflamed joints of infected mice, the presence of NK cells was also significantly elevated (Gardner et al., 2010). IL-12, which stimulates NK cell activity (Orange and Biron, 1996), was also present in high quantities, suggesting that activated NK cells play significant roles during CHIKV infection (Nakaya et al., 2012; Teo et al., 2015).
396
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
344 345 346 347 348 349 350 351 352 353 354 355 356 357
360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394
397 398 399 400 401 402
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 6 403 404 405 406 407 408 409 410 411 412 413 414 415 416
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
In clinical settings, it was previously reported that activated NK cells expressing CD69 were detected in the synovial fluid obtained from acutely infected CHIKF patients (Hoarau et al., 2010). Given that the functions of NK cells are tightly controlled by the concerted effects of activating and inhibitory receptors, it was revealed using PBMCs obtained from CHIKV-infected patients that a transient expansion of the highly cytotoxic NKG2C+ NK cells may function to decrease their own activity and limit overall immunopathology (Petitdemange et al., 2011). Nevertheless, the specific role of NK cells in CHIKV infection should be further explored with NK cell deficient animals, especially since NK cells adopted a pathogenic role in Venezuelan Equine Encephalitis virus (VEEV)-mediated central nervous system infection (Taylor et al., 2012).
417
12. Dendritic cells
418
435
Although rapid activation of plasmacytoid dendritic cells (pDCs) has been reported in patients during acute infection (Hoarau et al., 2010), little work has been done to understand the role of DCs. pDCs are major producers of Type I IFNs during viral infections (Siegal et al., 1999) that could initiate and activate NK cell mediated cytolysis (Colonna et al., 2004). However, CHIKV infection of pDCs isolated from healthy PMBCs did not secrete detectable amounts of both IFNa and IFNb (Schilte et al., 2010). More recently, the dendritic cell immunoreceptor (DCIR), a type of C-type leptin receptor (CLR), was demonstrated to play an important role in host protection against CHIKV infection in DCIR / mice (Long et al., 2013). This is in line with earlier studies where DCIR was reported to be one of the few CLRs that contain an intracellular immune receptor tyrosine-based inhibition motif (ITIM) (Meyer-Wentrup et al., 2008). Given that DCIR is expressed across other immune subsets such as monocytes and B cells (Bates et al., 1999), efforts to elucidate its role in these immune cells and in CHIKV immunopathology should be emphasized.
436
13. T cells
437
While innate immunity plays a role in the early phase of CHIKV infection, adaptive immunity comes in later for prolonged virus-specific memory responses (McCance and Huether, 2014). There are limited studies on the role of T cells in patients, except that CD8+ T cells dominate the early stages of the disease, with CD4+ T cells appearing at a later time to aid in the production of CHIKV-specific humoral response (Wauquier et al., 2011). However, it has also been reported that CD4+ T cells, but not CD8+ T cells were detected in the synovium of a patient suffering from chronic inflammation (Hoarau et al., 2010). To further decipher the roles of the adaptive immune response, RAG2 / mice were used (Teo et al., 2013). Interestingly, these immune-compromised animals did not exhibit any CHIKV-associated joint footpad pathology upon infection despite persistent viremia. However, a pathogenic role was elucidated for CD4+ T cells in driving joint inflammation when CD4 / animals were utilized (Teo et al., 2013). Cytokine profiling further indicated the involvement of a Th1-related pathway (Gardner et al., 2010; Nakaya et al., 2012; Teo et al., 2013, 2015). The pathogenic role of CD4+ T cells was further supported by another study where vaccination of B cell deficient lMT mice with whole inactivated virus was able to prime and activate CD4+ T cells to a level comparable to WT animals (Poo et al., 2014b). Subsequent challenge in these animals with CHIKV resulted in exacerbated joint footpad pathology, further confirming the pathogenic role of CD4+ T cells, albeit in a B cell deficient environment. Intriguingly, T cells were demonstrated to be protective in the
419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434
438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463
mouse models of other alphaviruses such as RRV and VEEV (Soden et al., 2000; Yun et al., 2009). Currently, the role of CD8+ T cells in CHIKV infection remains to be determined, although it has been shown in CD8 / mice that CD8+ T cells could be redundant in CHIKV infection (Teo et al., 2013). IL-17, a cytokine produced by Th17 T cells, has been detected in several cohorts of CHIKF patients (Ng et al., 2009; Chow et al., 2011; Teng et al., 2015). Th17 cells have been shown to play pathogenic roles in other alphaviral infections (Kulcsar et al., 2014), and in rheumatoid arthritis (RA) patients (Kotake et al., 1999; Chabaud et al., 2000). Given that the disease outcome of RA is quite similar to CHIKV infection (Nakaya et al., 2012), it would be interesting to elucidate the role of Th17 in defining CHIKV pathogenesis. It has been shown that a subpopulation of T cells called regulatory T cells (Tregs) is present to maintain immune tolerance, and Tregs are often studied in autoimmune diseases (Fessler et al., 2013; Dhaeze et al., 2015). But recent studies have shown increasing involvement of Tregs in infections (Belkaid and Rouse, 2005; Belkaid, 2007). More recently, Tregs were demonstrated in mice to protect against CHIKV-induced pathology by driving CHIKV-specific T cells into a state of anergy (Lee et al., 2015).
464
14. B cells
485
B cells and CHIKV-specific antibodies have been characterized extensively during the past 5 years (Kam et al., 2012a,b,c, 2014, 2015; Lum et al., 2013; Poo et al., 2014b). Studies using patient plasma samples showed that CHIKV infection induced a robust anti-CHIKV IgG antibody response dominated by the IgG3 isotype (Kam et al., 2012a). Further characterization of these anti-CHIKV antibodies led to the finding that viral load is a driving force in inducing the production of these neutralizing antibodies (Kam et al., 2012a). Likewise, in CHIKV-infected animals, high titers of neutralizing CHIKV-specific antibodies were produced after infection and their epitopes within the E2 glycoprotein has been mapped (Table 1). Furthermore, the virus-like particle (VLP) vaccine was demonstrated to induce the production of neutralizing antibodies in vaccinated non-human primates that protected the animals against challenge (Akahata et al., 2010). Likewise, a measles virus-based vaccine, which expresses CHIKV VLPs, protected susceptible mice from lethal challenge (Brandler et al., 2013). Recently, these promising vaccine candidates underwent small clinical trials, in which vaccinated healthy adults produced neutralizing antibodies, with no reports of adverse effects (Chang et al., 2014; Ramsauer et al., 2015). The specific role of CHIKV-specific antibodies in viremia clearance was also demonstrated using lMT mice, whereby viremia persisted in these animals for more than a year (Lum et al., 2013; Poo et al., 2014b). The importance of specific antibodies in resolving viremia was further proven by the passive transfer of human anti-CHIKV antibodies into IFNAR / or neonatal mice, which would otherwise succumb to infection (Couderc et al., 2009). It was further revealed that anti-CHIKV antibodies from patients and animals targeted a dominant epitope, E2EP3, which sits predominantly exposed on the CHIKV E2 glycoprotein (Kam et al., 2012b,c, 2014; Lum et al., 2013). The importance of protective antibodies was further demonstrated with the development of CHIKV-specific monoclonal antibodies (mAbs) from either mouse or human origin. These mAbs targeted epitopes located on CHIKV E2 (Warter et al., 2011; Fric et al., 2013; Pal et al., 2013; Selvarajah et al., 2013; Chua et al., 2014; Goh et al., 2015), E1 (Warter et al., 2011; Pal et al., 2013; Masrinoul et al., 2014), Capsid (Goh et al., 2015) and nsP2 (Chattopadhyay et al., 2014), and were protective against CHIKV infection in mice (Fric et al., 2013; Goh et al., 2013; Pal et al.,
486
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484
487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526
AVR 3649
No. of Pages 10, Model 5G
16 June 2015
Regions of epitopes found common to humans, mice and nonhuman primates are highlighted in bold type. The numbers correspond to the amino acid positions along the CHIKV viral genome. The first amino acid from nsP1 is annotated as 1. a
3121–3146 3177–3210 PTEGLEVTWGNNEPYKYWPQLSTNGT LLSMVGMAAGMCMCARRRCITPYELTPGATVPFL
7
2013; Selvarajah et al., 2013). However, it has also been shown that these protective antibodies were not necessarily CHIKV-isolate specific. In fact, some of these neutralizing antibodies have been shown to cross-react with closely related alphaviruses, such as O’Nyong Nyong virus (ONNV) and Sindbis virus (SINV) (Warter et al., 2011; Partidos et al., 2012; Goh et al., 2015). Despite their protective role, B cells and antibodies have also been suggested to enhance virus infections in a phenomenon known as antibody-dependent enhancement (ADE) (Takada and Kawaoka, 2003). ADE has been most well reported in Dengue virus (DENV) infections, whereby antibodies against a particular serotype of DENV can cross-react with another serotype, leading to an elevated level of infection and a more severe disease (Chareonsirisuthigul et al., 2007; Balsitis et al., 2010; Zellweger et al., 2010; Halstead, 2014). In DENV-infected patients, ADE can lead to severe dengue vascular permeability syndrome upon heterotypic DENV re-infection (Guzman et al., 2002; Alvarez et al., 2006; Guzman and Kouri, 2008). ADE is therefore a concern for CHIKV infections, especially with the emergence of possible quasi-species harboring mutations (Stapleford et al., 2014) that might render neutralizing antibodies enhancing at sub-neutralizing levels. In this scenario, severe disease due to re-infection could become a reality. In addition, CHIKV-specific antibodies have been suggested to be maternally transferred to their offspring (Watanaveeradej et al., 2006). Thus, while these maternally acquired antibodies are degraded with time, there exists a window during which these antibodies would become present in sub-neutralizing quantities, and expose the offspring to enhanced CHIKV infection, in a manner similar to DENV infection (Ng et al., 2014). Although an enhancement in the severity of disease was reported during a prime-boost immunization study (Hallengärd et al., 2014b), it remains to be determined if ADE can occur in CHIKV infections.
527
15. Future perspectives
560
Despite numerous studies, defining the precise functional role of immune cells in CHIKV-infected humans remains a challenge. Researchers could further explore alternative approaches to better understand immunopathogenesis. Among the approaches, the development of viable mouse models has assisted in addressing some questions. However, as the immune systems of mice and humans are not identical, new research tools must be designed to aid in future studies. The development of humanized mice (Shultz et al., 2007) with immune cells of human origin may offer a possible research direction to define immunopathogenesis. The dream of having a chikungunya vaccine could be near with the success of the VLP vaccine (Chang et al., 2014). Nevertheless, caution should still be exercised during vaccine design, so that vaccines generated do not prime a sub-par humoral response, which might lead to an exacerbated disease and pathology in patients. Likewise, given the proposed pathogenic roles of CD4+ T cells, vaccine formulations targeting T cells could be further manipulated to prime protective CD4+ T cells. In addition, given that both monocytes and CD4+ T cells are potentially pathogenic during CHIKV infection, drugs targeting these subsets could be explored. Furthermore, with Tregs shown to play an important immune-regulatory role, it would be interesting to explore whether Tregs could also serve as a plausible point of intervention.
561
16. Uncited references
584
Lumsden (1955) and Rezza et al. (2007).
528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559
562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583
585
b
3177–3210
3097–3146 3073–3098
Region on E2 glycoprotein
Part of the acid-sensitive region 2985–3002 3025–3066 GNVKITVNGQTVRYKCNC HAAVTNHKKWQYNSPLVPRNAE LGDRKGKIHIPFPLANVTCR 2985–3002 3025–3081
GNVKITVNGQTVRYKCNC HAAVTNHKKWQYNSPLVPRNAEL GDRKGKIHIPFPLANVTCRVPKARNPTVTYGKNQ RNMGEEPNYQEEWVMHKKEVVLT VPTEGLEVTWGNNEPYKYWPQLSTNGT LLSMVGMAAGMCMCARRRCITPYELTPGATVPFL 3009–3034 3025–3062
2800–2818 2961–2978
Amino acid Macaque B cell epitopes
STKDNFNVYKATRPYLAHC ATTEEIEVHMPPDTPDRT 2800–2818 2929–2954
Amino acid Mouse B cell epitopes
STKDNFNVYKATRPYLAHC HHDPPVIGREKFHSRPQHGKELPCST 2800–2818 2841–2882
STKDNFNVYKATRPYLAHC TDGTLKIQVSLQIGIKTDDSH DWTKLRYMDNHMPADAERAGL LTTTDKVINNCKVDQCHAAVTNHKKW HAAVTNHKKWQYNSPLVPRNA ELGDRKGKIHIPFPLAN PTVTYGKNQVIMLLYPDHPTLLSYRN
Amino acidb Human B cell epitopesa
Table 1 Comparison of identified CHIKV B cell epitopes in humans, mice and macaques, within the E2 proteome.
E2EP3 region
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
586
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 8
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
587
Acknowledgements
588
591
We acknowledge Wendy Lee and Kai Er Eng from SIgN for their critical comments of this manuscript. Fok-Moon Lum is supported by the President Graduate Fellowship from the Yong Loo Lin School of Medicine, National University of Singapore (Singapore).
592
References
593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665
Akahata, W., Yang, Z.Y., Andersen, H., Sun, S., Holdaway, H.A., Kong, W.P., Lewis, M.G., Higgs, S., Rossmann, M.G., Rao, S., Nabel, G.J., 2010. A virus-like particle vaccine for epidemic chikungunya virus protects nonhuman primates against infection. Nat. Med. 16 (3), 334–338. Alvarez, M., Rodriguez-Roche, R., Bernardo, L., Vazquez, S., Morier, L., Gonzalez, D., Castro, O., Kouri, G., Halstead, S.B., Guzman, M.G., 2006. Dengue hemorrhagic fever caused by sequential dengue 1–3 virus infections over a long time interval: Havana epidemic, 2001–2002. Am. J. Trop. Med. Hyg. 75 (6), 1113– 1117. Balsitis, S.J., Williams, K.L., Lachica, R., Flores, D., Kyle, J.L., Mehlhop, E., Johnson, S., Diamond, M.S., Beatty, P.R., Harris, E., 2010. Lethal antibody enhancement of dengue disease in mice is prevented by fc modification. PLoS Pathog. 6 (2), e1000790. Bates, E.E., Fournier, N., Garcia, E., Valladeau, J., Durand, I., Pin, J.J., Zurawski, S.M., Patel, S., Abrams, J.S., Lebecque, S., 1999. APCS express DCIR, a novel c-type lectin surface receptor containing an immunoreceptor tyrosine-based inhibitory motif. J. Immunol. 163 (4), 1973–1983. Belkaid, Y., 2007. Regulatory t cells and infection: a dangerous necessity. Nat. Rev. Immunol. 7 (11), 875–888. Belkaid, Y., Rouse, B.T., 2005. Natural regulatory t cells in infectious disease. Nat. Immunol. 6 (4), 353–360. Bhatia, M., Ramnath, R.D., Chevali, L., Guglielmotti, A., 2005. Treatment with Bindarit, a blocker of mcp-1 synthesis, protects mice against acute pancreatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 288 (6), G1259–G1265. Borgherini, G., Poubeau, P., Staikowsky, F., Lory, M., le Moullec, N., Becquart, J.P., Wengling, C., Michault, A., Paganin, F., 2007. Outbreak of chikungunya on reunion island: early clinical and laboratory features in 157 adult patients. Clin. Infect. Dis. 44 (11), 1401–1407. Brandler, S., Ruffie, C., Combredet, C., Brault, J.B., Najburg, V., Prevost, M.C., Habel, A., Tauber, E., Despres, P., Tangy, F., 2013. A recombinant measles vaccine expressing chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine 31 (36), 3718–3725. Bréhin, A.C., Casadémont, I., Frenkiel, M.P., Julier, C., Sakuntabhai, A., Desprès, P., 2009. The large form of human 20 ,50 -oligoadenylate synthetase (oas3) exerts antiviral effect against chikungunya virus. Virology 384 (1), 216–222. Chabaud, M., Garnero, P., Dayer, J.M., Guerne, P.A., Fossiez, F., Miossec, P., 2000. Contribution of interleukin 17 to synovium matrix destruction in rheumatoid arthritis. Cytokine 12 (7), 1092–1099. Chandak, N.H., Kashyap, R.S., Kabra, D., Karandikar, P., Saha, S.S., Morey, S.H., Purohit, H.J., Taori, G.M., Daginawala, H.F., 2009. Neurological complications of chikungunya virus infection. Neurol. India 57 (2), 177–180. Chang, L.J., Dowd, K.A., Mendoza, F.H., Saunders, J.G., Sitar, S., Plummer, S.H., Yamshchikov, G., Sarwar, U.N., Hu, Z., Enama, M.E., Bailer, R.T., Koup, R.A., Schwartz, R.M., Akahata, W., Nabel, G.J., Mascola, J.R., Pierson, T.C., Graham, B.S., Ledgerwood, J.E., Team, V.R.C.S., 2014. Safety and tolerability of chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet 384 (9959), 2046–2052. Chareonsirisuthigul, T., Kalayanarooj, S., 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 proinflammatory cytokine production, in thp-1 cells. J. Gen. Virol. 88 (2), 365–375. Chattopadhyay, S., Kumar, A., Mamidi, P., Nayak, T.K., Das, I., Chhatai, J., Basantray, I., Bramha, U., Maiti, P.K., Singh, S., Suryawanshi, A.R., Chattopadhyay, S., 2014. Development and characterization of monoclonal antibody against nonstructural protein-2 of chikungunya virus and its application. J. Virol. Methods 199, 86–94. Chen, C.I., Clark, D.C., Pesavento, P., Lerche, N.W., Luciw, P.A., Reisen, W.K., Brault, A.C., 2010. Comparative pathogenesis of epidemic and enzootic chikungunya viruses in a pregnant rhesus macaque model. Am. J. Trop. Med. Hyg. 83 (6), 1249–1258. Chen, W., Foo, S.S., Rulli, N.E., Taylor, A., Sheng, K.C., Herrero, L.J., Herring, B.L., Lidbury, B.A., Li, R.W., Walsh, N.C., Sims, N.A., Smith, P.N., Mahalingam, S., 2014. Arthritogenic alphaviral infection perturbs osteoblast function and triggers pathologic bone loss. Proc. Natl. Acad. Sci. U. S. A. 111 (16), 6040–6045. Chen, W., Foo, S.S., Taylor, A., Lulla, A., Merits, A., Hueston, L., Forwood, M.R., Walsh, N.C., Sims, N.A., Herrero, L.J., Mahalingam, S., 2015. Bindarit, an inhibitor of monocyte chemotactic protein synthesis, protects against bone loss induced by chikungunya virus infection. J. Virol. 89 (1), 581–593. Chow, A., Her, Z., Ong, E.K.S., Chen, J.M., Dimatatac, F., Kwek, D.J.C., Barkham, T., Yang, H., Rénia, L., Leo, Y.S., Ng, L.F.P., 2011. Persistent arthralgia induced by chikungunya virus infection is associated with interleukin-6 and granulocyte macrophage colony-stimulating factor. J. Infect. Dis. 203 (2), 149–157.
589 590
Chua, C.L., Chan, Y.F., Sam, I.C., 2014. Characterisation of mouse monoclonal antibodies targeting linear epitopes on chikungunya virus e2 glycoprotein. J. Virol. Methods 195, 126–133. Clavarino, G., Claudio, N., Couderc, T., Dalet, A., Judith, D., Camosseto, V., Schmidt, E.K., Wenger, T., Lecuit, M., Gatti, E., Pierre, P., 2012. Induction of GADD34 is necessary for dsRNA-dependent interferon-beta production and participates in the control of chikungunya virus infection. PLoS Pathog. 8 (5), e1002708. Colonna, M., Trinchieri, G., Liu, Y.J., 2004. Plasmacytoid dendritic cells in immunity. Nat. Immunol. 5 (12), 1219–1226. Couderc, T., Chrétien, F., Schilte, C., Disson, O., Brigitte, M., Guivel-Benhassine, F., Touret, Y., Barau, G., Cayet, N., Schuffenecker, I., Desprès, P., ArenzanaSeisdedos, F., Michault, A., Albert, M.L., Lecuit, M., 2008. A mouse model for chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog. 4 (2), e29. Couderc, T., Khandoudi, N., Grandadam, M., Visse, C., Gangneux, N., Bagot, S., Prost, J.F., Lecuit, M., 2009. Prophylaxis and therapy for chikungunya virus infection. J. Infect. Dis. 200 (4), 516–523. Decker, T., Stockinger, S., Karaghiosoff, M., Muller, M., Kovarik, P., 2002. IFNS and stats in innate immunity to microorganisms. J. Clin. Invest. 109 (10), 1271– 1277. Dhaeze, T., Stinissen, P., Liston, A., Hellings, N., 2015. Humoral autoimmunity: a failure of regulatory t cells? Autoimmun. Rev. http://dx.doi.org/10.1016/ j.autrev.2015.04.006 [epub ahead of print]. Economopoulou, A., Dominguez, M., Helynck, B., Sissoko, D., Wichmann, O., Quenel, P., Germonneau, P., Quatresous, I., 2009. Atypical chikungunya virus infections: clinical manifestations, mortality and risk factors for severe disease during the 2005–2006 outbreak on reunion. Epidemiol. Infect. 137 (4), 534–541. Fessler, J., Felber, A., Duftner, C., Dejaco, C., 2013. Therapeutic potential of regulatory t cells in autoimmune disorders. BioDrugs 27 (4), 281–291. Fric, J., Bertin-Maghit, S., Wang, C.I., Nardin, A., Warter, L., 2013. Use of human monoclonal antibodies to treat chikungunya virus infection. J. Infect. Dis. 207 (2), 319–322. Fros, J.J., Liu, W.J., Prow, N.A., Geertsema, C., Ligtenberg, M., Vanlandingham, D.L., Schnettler, E., Vlak, J.M., Suhrbier, A., Khromykh, A.A., Pijlman, G.P., 2010. Chikungunya virus nonstructural protein 2 inhibits type I/II interferonstimulated JAK-STAT signaling. J. Virol. 84 (20), 10877–10887. Gardner, J., Anraku, I., Le, T.T., Larcher, T., Major, L., Roques, P., Schroder, W.A., Higgs, S., Suhrbier, A., 2010. Chikungunya virus arthritis in adult wild-type mice. J. Virol. 84 (16), 8021–8032. Glick, D., Barth, S., Macleod, K.F., 2010. Autophagy: cellular and molecular mechanisms. J. Pathol. 221 (1), 3–12. Goh, L.Y., Hobson-Peters, J., Prow, N.A., Gardner, J., Bielefeldt-Ohmann, H., Pyke, A.T., Suhrbier, A., Hall, R.A., 2013. Neutralizing monoclonal antibodies to the e2 protein of chikungunya virus protects against disease in a mouse model. Clin. Immunol. 149 (3), 487–497. Goh, L.Y., Hobson-Peters, J., Prow, N.A., Gardner, J., Bielefeldt-Ohmann, H., Suhrbier, A., Hall, R.A., 2015. Monoclonal antibodies specific for the capsid protein of chikungunya virus suitable for multiple applications. J. Gen. Virol. 96 (3), 507– 512. Guzman, M.G., Kouri, G., 2008. Dengue haemorrhagic fever integral hypothesis: confirming observations, 1987–2007. Trans. R. Soc. Trop. Med. Hyg. 102 (6), 522–523. Guzman, M.G., Kouri, G., Valdes, L., Bravo, J., Vazquez, S., Halstead, S.B., 2002. Enhanced severity of secondary dengue-2 infections: death rates in 1981 and 1997 cuban outbreaks. Rev. Panam. Salud Publica 11 (4), 223–227. Hallengärd, D., Lum, F.M., Kümmerer, B.M., Lulla, A., Lulla, V., García-Arriaza, J., Fazakerley, J.K., Roques, P., le Grand, R., Merits, A., Ng, L.F.P., Esteban, M., Liljeström, P., 2014. Prime-boost immunization strategies against chikungunya virus. J. Virol. 88 (22), 13333–13343. Halstead, S.B., 2014. Dengue antibody-dependent enhancement: knowns and unknowns. Microbiol. Spec. 2 (6). Her, Z., Kam, Y.W., Lin, R.T.P., Ng, L.F.P., 2009. Chikungunya: a bending reality. Microbes Infect. 11 (14), 1165–1176. Her, Z., Malleret, B., Chan, M., Ong, E.K.S., Wong, S.C., Kwek, D.J.C., Tolou, H., Lin, R.T.P., Tambyah, P.A., Rénia, L., Ng, L.F.P., 2010. Active infection of human blood monocytes by chikungunya virus triggers an innate immune response. J. Immunol. 184 (10), 5903–5913. Her, Z., Teng, T.S., Tan, J.J., Teo, T.H., Kam, Y.W., Lum, F.M., Lee, W.W., Gabriel, C., Melchiotti, R., Andiappan, A.K., Lulla, V., Lulla, A., Win, M.K., Chow, A., Biswas, S.K., Leo, Y.S., Lecuit, M., Merits, A., Renia, L., Ng, L.F., 2015. Loss of TLR3 aggravates CHIKV replication and pathology due to an altered virus-specific neutralizing antibody response. EMBO Mol. Med. 7 (1), 24–41. Herrero, L.J., Nelson, M., Srikiatkhachorn, A., Gu, R., Anantapreecha, S., FingerleRowson, G., Bucala, R., Morand, E., Santos, L.L., Mahalingam, S., 2011. Critical role for macrophage migration inhibitory factor (MIF) in Ross River virusinduced arthritis and myositis. Proc. Natl. Acad. Sci. U. S. A. 108 (29), 12048– 12053. Herrero, L.J., Sheng, K.C., Jian, P., Taylor, A., Her, Z., Herring, B.L., Chow, A., Leo, Y.S., Hickey, M.J., Morand, E.F., Ng, L.F., Bucala, R., Mahalingam, S., 2013. Macrophage migration inhibitory factor receptor CD74 mediates alphavirus-induced arthritis and myositis in murine models of alphavirus infection. Arthritis Rheum. 65 (10), 2724–2736. Hoarau, J.J., Jaffar Bandjee, M.C., Krejbich Trotot, P., Das, T., Li-Pat-Yuen, G., Dassa, B., Denizot, M., Guichard, E., Ribera, A., Henni, T., Tallet, F., Moiton, M.P., Gauzère, B.A., Bruniquet, S., Jaffar Bandjee, Z., Morbidelli, P., Martigny, G., Jolivet, M., Gay, F., Grandadam, M., Tolou, H., Vieillard, V., Debré, P., Autran, B., Gasque, P., 2010.
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837
Persistent chronic inflammation and infection by chikungunya arthritogenic alphavirus in spite of a robust host immune response. J. Immunol. 184 (10), 5914–5927. Hofbauer, L.C., Khosla, S., Dunstan, C.R., Lacey, D.L., Spelsberg, T.C., Riggs, B.L., 1999. Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 140 (9), 4367–4370. Honda, K., Takaoka, A., Taniguchi, T., 2006. Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 25 (3), 349–360. Janeway Jr., C.A., Medzhitov, R., 2002. Innate immune recognition. Annu. Rev. Immunol. 20, 197–216. Jones, P.H., Maric, M., Madison, M.N., Maury, W., Roller, R.J., Okeoma, C.M., 2013. BST-2/tetherin-mediated restriction of chikungunya (CHIKV) VLP budding is counteracted by CHIKV non-structural protein 1 (NSP1). Virology 438 (1), 37– 49. Joubert, P.E., Werneke, S.W., de la Calle, C., Guivel-Benhassine, F., Giodini, A., Peduto, L., Levin, B., Schwartz, O., Lenschow, D.J., Albert, M.L., 2012. Chikungunya virusinduced autophagy delays caspase-dependent cell death. J. Exp. Med. 209 (5), 1029–1047. Judith, D., Mostowy, S., Bourai, M., Gangneux, N., Lelek, M., Lucas-Hourani, M., Cayet, N., Jacob, Y., Prevost, M.C., Pierre, P., Tangy, F., Zimmer, C., Vidalain, P.O., Couderc, T., Lecuit, M., 2013. Species-specific impact of the autophagy machinery on chikungunya virus infection. EMBO Rep. 14 (6), 534–544. Kam, Y.W., Lee, W.W.L., Simarmata, D., Harjanto, S., Teng, T.S., Tolou, H., Chow, A., Lin, R.T.P., Leo, Y.-S., Rénia, L., Ng, L.F.P., 2012c. Longitudinal analysis of the human antibody response to chikungunya virus infection: implications for serodiagnosis assays and vaccine development. J. Virol. 86 (23), 13005–13015. Kam, Y.W., Lee, W.W.L., Simarmata, D., le Grand, R., Tolou, H., Merits, A., Roques, P., Ng, L.F., 2014. Unique epitopes recognized by antibodies induced in chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS One 9 (4), e95647. Kam, Y.W., Lum, F.M., Teo, T.H., Lee, W.W.L., Simarmata, D., Harjanto, S., Chua, C.L., Chan, Y.F., Wee, J.K., Chow, A., Lin, R.T.P., Leo, Y.S., le Grand, R., Sam, I.C., Tong, J.C., Roques, P., Wiesmüller, K.H., Rénia, L., Rötzschke, O., Ng, L.F.P., 2012b. Early neutralizing igg response to chikungunya virus in infected patients targets a dominant linear epitope on the e2 glycoprotein. EMBO Mol. Med. 4 (4), 330– 343. Kam, Y.W., Ong, E.K., Rénia, L., Tong, J.C., Ng, L.F., 2009. Immuno-biology of chikungunya and implications for disease intervention. Microbes Infect. 11 (14), 1186–1196. Kam, Y.W., Simarmata, D., Chow, A., Her, Z., Teng, T.S., Ong, E.K.S., Rénia, L., Leo, Y.S., Ng, L.F.P., 2012a. Early appearance of neutralizing immunoglobulin g3 antibodies is associated with chikungunya virus clearance and long-term clinical protection. J. Infect. Dis. 205 (7), 1147–1154. Kam, Y.W., Pok, K.Y., Eng, K.E., Tan, L.K., Kaur, S., Lee, W.W.L., Leo, Y.S., Ng, L.C., Ng, L.F., 2015. Sero-prevalence and cross-reactivity of chikungunya virus specific anti-e2ep3 antibodies in arbovirus-infected patients. PLoS Negl. Trop. Dis. 9 (1), e3445. Kashyap, R.S., Morey, S.H., Chandak, N.H., Purohit, H.J., Taori, G.M., Daginawala, H.F., 2010. Detection of viral antigen, IGM and IGG antibodies in cerebrospinal fluid of chikungunya patients with neurological complications. Cerebrospinal Fluid Res. 7, 12. Kawai, T., Akira, S., 2010. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat. Immunol. 11 (5), 373–384. Kawai, T., Takahashi, K., Sato, S., Coban, C., Kumar, H., Kato, H., Ishii, K.J., Takeuchi, O., Akira, S., 2005. IPS-1, an adaptor triggering RIG-I- and MDA5-mediated type I interferon induction. Nat. Immunol. 6 (10), 981–988. Kelvin, A.A., Banner, D., Silvi, G., Moro, M.L., Spataro, N., Gaibani, P., Cavrini, F., Pierro, A., Rossini, G., Cameron, M.J., Bermejo-Martin, J.F., Paquette, S.G., Xu, L., Danesh, A., Farooqui, A., Borghetto, I., Kelvin, D.J., Sambri, V., Rubino, S., 2011. Inflammatory cytokine expression is associated with chikungunya virus resolution and symptom severity. PLoS Negl. Trop. Dis. 5 (8), e1279. Kotake, S., Udagawa, N., Takahashi, N., Matsuzaki, K., Itoh, K., Ishiyama, S., Saito, S., Inoue, K., Kamatani, N., Gillespie, M.T., Martin, T.J., Suda, T., 1999. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J. Clin. Invest. 103 (9), 1345–1352. Krejbich-Trotot, P., Denizot, M., Hoarau, J.J., Jaffar-Bandjee, M.C., Das, T., Gasque, P., 2011a. Chikungunya virus mobilizes the apoptotic machinery to invade host cell defenses. FASEB J. 25 (1), 314–325. Krejbich-Trotot, P., Gay, B., Li-Pat-yuen, G., Hoarau, J.J., Jaffar-Bandjee, M.C., Briant, L., Gasque, P., Denizot, M., 2011b. Chikungunya triggers an autophagic process which promotes viral replication. Virol. J. 8 (1), 432. Kulcsar, K.A., Baxter, V.K., Greene, I.P., Griffin, D.E., 2014. Interleukin 10 modulation of pathogenic th17 cells during fatal alphavirus encephalomyelitis. Proc. Natl. Acad. Sci. U. S. A. 111 (45), 16053–16058. Kupper, T.S., Fuhlbrigge, R.C., 2004. Immune surveillance in the skin: mechanisms and clinical consequences. Nat. Rev. Immunol. 4 (3), 211–222. Labadie, K., Larcher, T., Joubert, C., Mannioui, A., Delache, B., Brochard, P., Guigand, L., Dubreil, L., Lebon, P., Verrier, B., de Lamballerie, X., Suhrbier, A., Cherel, Y., le Grand, R., Roques, P., 2010. Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J. Clin. Invest. 120 (3), 894–906. Lee, W.W.L., Teo, T.H., Her, Z., Lum, F.M., Kam, Y.W., Hasse, D., Rénia, L., Rötzschke, O., Ng, L.F.P., 2015. Expanding regulatory T cells alleviates chikungunya virusinduced pathology in mice. J. Virol. http://dx.doi.org/10.1128/JVI.00998-15 [epub ahead of print].
9
Leung, J.Y.S., Ng, M.M.L., Chu, J.J.H., 2011. Replication of alphaviruses: a review on the entry process of alphaviruses into cells. Adv. Virol. http://dx.doi.org/ 10.1155/2011/249640. Li, Y., Backesjo, C.M., Haldosen, L.A., Lindgren, U., 2008. IL-6 receptor expression and IL-6 effects change during osteoblast differentiation. Cytokine 43 (2), 165–173. Lidbury, B.A., Rulli, N.E., Suhrbier, A., Smith, P.N., McColl, S.R., Cunningham, A.L., Tarkowski, A., van Rooijen, N., Fraser, R.J., Mahalingam, S., 2008. Macrophagederived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J. Infect. Dis. 197 (11), 1585–1593. Lidbury, B.A., Simeonovic, C., Maxwell, G.E., Marshall, I.D., Hapel, A.J., 2000. Macrophage-induced muscle pathology results in morbidity and mortality for Ross River virus-infected mice. J. Infect. Dis. 181 (1), 27–34. Long, K.M., Whitmore, A.C., Ferris, M.T., Sempowski, G.D., McGee, C., Trollinger, B., Gunn, B., Heise, M.T., 2013. Dendritic cell immunoreceptor regulates chikungunya virus pathogenesis in mice. J. Virol. 87 (10), 5697–5706. Long, K.M., Heise, M.T., 2015. Protective and pathogenic responses to chikungunya virus infection. Curr. Trop. Med. Rep. 2, 13–21. Lu, B., Rutledge, B.J., Gu, L., Fiorillo, J., Lukacs, N.W., Kunkel, S.L., North, R., Gerard, C., Rollins, B.J., 1998. Abnormalities in monocyte recruitment and cytokine expression in monocyte chemoattractant protein 1-deficient mice. J. Exp. Med. 187 (4), 601–608. Lum, F.M., Teo, T.H., Lee, W.W.L., Kam, Y.W., Rénia, L., Ng, L.F.P., 2013. An essential role of antibodies in the control of chikungunya virus infection. J. Immunol. 190 (12), 6295–6302. Lumsden, W.H., 1955. An epidemic of virus disease in Southern Province, Tanganyika Territory, in 1952–53. II. General description and epidemiology. Trans. R. Soc. Trop. Med. Hyg. 49 (1), 33–57. Masrinoul, P., Puiprom, O., Tanaka, A., Kuwahara, M., Chaichana, P., Ikuta, K., Ramasoota, P., Okabayashi, T., 2014. Monoclonal antibody targeting chikungunya virus envelope 1 protein inhibits virus release. Virology 464– 465, 111–117. Mccance, K.L., Huether, S.E., 2014. Pathophysiology: The Biologic Basis for Disease in Adults and Children. Elsevier Health Sciences. Medzhitov, R., Janeway, C., 2000. Innate immune recognition: mechanisms and pathways. Immunol. Rev. 173 (1), 89–97. Melton, J.V., Ewart, G.D., Weir, R.C., Board, P.G., Lee, E., Gage, P.W., 2002. Alphavirus 6K proteins form ion channels. J. Biol. Chem. 277 (49), 46923–46931. Messaoudi, I., Vomaske, J., Totonchy, T., Kreklywich, C.N., Haberthur, K., Springgay, L., Brien, J.D., Diamond, M.S., Defilippis, V.R., Streblow, D.N., 2013. Chikungunya virus infection results in higher and persistent viral replication in aged rhesus macaques due to defects in anti-viral immunity. PLoS Negl. Trop. Dis. 7 (7), e2343. Meyer-Wentrup, F., Benitez-Ribas, D., Tacken, P.J., Punt, C.J., Figdor, C.G., de Vries, I.J., Adema, G.J., 2008. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood 111 (8), 4245–4253. Nakaya, H.I., Gardner, J., Poo, Y.S., Major, L., Pulendran, B., Suhrbier, A., 2012. Gene profiling of chikungunya virus arthritis in a mouse model reveals significant overlap with rheumatoid arthritis. Arthritis Rheum. 64 (11), 3553–3563. Ng, J.K., Zhang, S.L., Tan, H.C., Yan, B., Martinez, J.M., Tan, W.Y., Lam, J.H., Tan, G.K., Ooi, E.E., Alonso, S., 2014. First experimental in vivo model of enhanced dengue disease severity through maternally acquired heterotypic dengue antibodies. PLoS Pathog. 10 (4), e1004031. Ng, L.F.P., Chow, A., Sun, Y.-J., Kwek, D.J.C., Lim, P.-L., Dimatatac, F., Ng, L.-C., Ooi, E.E., Choo, K.-H., Her, Z., Kourilsky, P., Leo, Y.-S., 2009. IL-1beta, IL-6, and rantes as biomarkers of chikungunya severity. PLoS One 4 (1), e4261. Noret, M., Herrero, L., Rulli, N., Rolph, M., Smith, P.N., Li, R.W., Roques, P., Gras, G., Mahalingam, S., 2012. Interleukin 6, rankl, and osteoprotegerin expression by chikungunya virus-infected human osteoblasts. J. Infect. Dis. 206 (3), 455–457: 457–459. Ong, R.Y., Lum, F.M., Ng, L.F., 2014. The fine line between protection and pathology in neurotropic flavivirus and alphavirus infections. Fut. Virol. 9 (3), 313–330. Orange, J.S., Biron, C.A., 1996. An absolute and restricted requirement for IL-12 in natural killer cell IFN-gamma production and antiviral defense. Studies of natural killer and T cell responses in contrasting viral infections. J. Immunol. 156 (3), 1138–1142. Ozden, S., Huerre, M., Riviere, J.P., Coffey, L.L., Afonso, P.V., Mouly, V., de Monredon, J., Roger, J.C., el Amrani, M., Yvin, J.L., 2007. Human muscle satellite cells as targets of chikungunya virus infection. PLoS One 2 (6), e527. Pal, P., Dowd, K.A., Brien, J.D., Edeling, M.A., Gorlatov, S., Johnson, S., Lee, I., Akahata, W., Nabel, G.J., Richter, M.K., Smit, J.M., Fremont, D.H., Pierson, T.C., Heise, M.T., Diamond, M.S., 2013. Development of a highly protective combination monoclonal antibody therapy against chikungunya virus. PLoS Pathog. 9 (4), e1003312. Panning, M., Grywna, K., van Esbroeck, M., Emmerich, P., Drosten, C., 2008. Chikungunya fever in travelers returning to Europe from the Indian Ocean Region, 2006. Emerg. Infect. Dis. 14 (3), 416–422. Partidos, C.D., Paykel, J., Weger, J., Borland, E.M., Powers, A.M., Seymour, R., Weaver, S.C., Stinchcomb, D.T., Osorio, J.E., 2012. Cross-protective immunity against O’nyong–Nyong virus afforded by a novel recombinant chikungunya vaccine. Vaccine 30 (31), 4638–4643. Petitdemange, C., Becquart, P., Wauquier, N., Béziat, V., Debré, P., Leroy, E.M., Vieillard, V., 2011. Unconventional repertoire profile is imprinted during acute chikungunya infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog. 7 (9), e1002268.
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009
838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923
AVR 3649
No. of Pages 10, Model 5G
16 June 2015 10 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999
F.-M. Lum, L.F.P. Ng / Antiviral Research xxx (2015) xxx–xxx
Poo, Y.S., Nakaya, H., Gardner, J., Larcher, T., Schroder, W.A., Le, T.T., Major, L.D., Suhrbier, A., 2014a. CCR2 deficiency promotes exacerbated chronic erosive neutrophil-dominated chikungunya virus arthritis. J. Virol. 88 (12), 6862–6872. Poo, Y.S., Rudd, P.A., Gardner, J., Wilson, J.A., Larcher, T., Colle, M.A., Le, T.T., Nakaya, H.I., Warrilow, D., Allcock, R., Bielefeldt-Ohmann, H., Schroder, W.A., Khromykh, A.A., Lopez, J.A., Suhrbier, A., 2014b. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl. Trop. Dis. 8 (12), e3354. Ramsauer, K., Schwameis, M., Firbas, C., Mullner, M., Putnak, R.J., Thomas, S.J., Despres, P., Tauber, E., Jilma, B., Tangy, F., 2015. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect. Dis. 15 (5), 519–527. Rashad, A.A., Mahalingam, S., Keller, P.A., 2014. Chikungunya virus: emerging targets and new opportunities for medicinal chemistry. J. Med. Chem. 57 (4), 1147–1166. Rezza, G., Nicoletti, L., Angelini, R., Romi, R., Finarelli, A.C., Panning, M., Cordioli, P., Fortuna, C., Boros, S., Magurano, F., Silvi, G., Angelini, P., Dottori, M., Ciufolini, M.G., Majori, G.C., Cassone, A., Group, C.S., 2007. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet 370 (9602), 1840–1846. Romano, M., Sironi, M., Toniatti, C., Polentarutti, N., Fruscella, P., Ghezzi, P., Faggioni, R., Luini, W., van Hinsbergh, V., Sozzani, S., 1997. Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment. Immunity 6 (3), 315–325. Rudd, P.A., Wilson, J., Gardner, J., Larcher, T., Babarit, C., Le, T.T., Anraku, I., Kumagai, Y., Loo, Y.M., Gale Jr., M., Akira, S., Khromykh, A.A., Suhrbier, A., 2012. Interferon response factors 3 and 7 protect against chikungunya virus hemorrhagic fever and shock. J. Virol. 86 (18), 9888–9898. Rulli, N.E., Rolph, M.S., Srikiatkhachorn, A., Anantapreecha, S., Guglielmotti, A., Mahalingam, S., 2011. Protection from arthritis and myositis in a mouse model of acute chikungunya virus disease by bindarit, an inhibitor of monocyte chemotactic protein-1 synthesis. J. Infect. Dis. 204 (7), 1026–1030. Salvador, B., Zhou, Y., Michault, A., Muench, M.O., Simmons, G., 2009. Characterization of chikungunya pseudotyped viruses: identification of refractory cell lines and demonstration of cellular tropism differences mediated by mutations in e1 glycoprotein. Virology 393 (1), 33–41. Schilte, C., Buckwalter, M.R., Laird, M.E., Diamond, M.S., Schwartz, O., Albert, M.L., 2012. Cutting edge: independent roles for IRF-3 and IRF-7 in hematopoietic and nonhematopoietic cells during host response to chikungunya infection. J. Immunol. 188 (7), 2967–2971. Schilte, C., Couderc, T., Chretien, F., Sourisseau, M., Gangneux, N., Guivel-Benhassine, F., Kraxner, A., Tschopp, J., Higgs, S., Michault, A., Arenzana-Seisdedos, F., Colonna, M., Peduto, L., Schwartz, O., Lecuit, M., Albert, M.L., 2010. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J. Exp. Med. 207 (2), 429–442. Schoggins, J.W., Rice, C.M., 2011. Interferon-stimulated genes and their antiviral effector functions. Curr. Opin. Virol. 1 (6), 519–525. Schoggins, J.W., Wilson, S.J., Panis, M., Murphy, M.Y., Jones, C.T., Bieniasz, P., Rice, C.M., 2011. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 472 (7344), 481–485. Schwartz, O., Albert, M.L., 2010. Biology and pathogenesis of chikungunya virus. Nat. Rev. Microbiol. 8 (7), 491–500. Selvarajah, S., Sexton, N.R., Kahle, K.M., Fong, R.H., Mattia, K.A., Gardner, J., Lu, K., Liss, N.M., Salvador, B., Tucker, D.F., Barnes, T., Mabila, M., Zhou, X., Rossini, G., Rucker, J.B., Sanders, D.A., Suhrbier, A., Sambri, V., Michault, A., Muench, M.O., Doranz, B.J., Simmons, G., 2013. A neutralizing monoclonal antibody targeting the acid-sensitive region in chikungunya virus e2 protects from disease. PLoS Negl. Trop. Dis. 7 (9), e2423. Shultz, L.D., Ishikawa, F., Greiner, D.L., 2007. Humanized mice in translational biomedical research. Nat. Rev. Immunol. 7 (2), 118–130. Siegal, F.P., Kadowaki, N., Shodell, M., Fitzgerald-Bocarsly, P.A., Shah, K., Ho, S., Antonenko, S., Liu, Y.-J., 1999. The nature of the principal type 1 interferonproducing cells in human blood. Science 284 (5421), 1835–1837. Snyder, A.J., Mukhopadhyay, S., 2012. The alphavirus e3 glycoprotein functions in a clade-specific manner. J. Virol. 86 (24), 13609–13620. Soden, M., Vasudevan, H., Roberts, B., Coelen, R., Hamlin, G., Vasudevan, S., la Brooy, J., 2000. Detection of viral ribonucleic acid and histologic analysis of inflamed synovium in Ross River virus infection. Arthritis Rheum. 43 (2), 365–369. Solignat, M., Gay, B., Higgs, S., Briant, L., Devaux, C., 2009. Replication cycle of chikungunya: a re-emerging arbovirus. Virology 393 (2), 183–197. Sourisseau, M., Schilte, C., Casartelli, N., Trouillet, C., Guivel-Benhassine, F., Rudnicka, D., Sol-Foulon, N., le Roux, K., Prevost, M.C., Fsihi, H., Frenkiel, M.P., Blanchet, F., Afonso, P.V., Ceccaldi, P.E., Ozden, S., Gessain, A., Schuffenecker, I., Verhasselt, B., Zamborlini, A., Saib, A., Rey, F.A., Arenzana-Seisdedos, F., Despres, P., Michault, A., Albert, M.L., Schwartz, O., 2007. Characterization of reemerging chikungunya virus. PLoS Pathog. 3 (6), e89.
Stapleford, K.A., Coffey, L.L., Lay, S., Borderia, A.V., Duong, V., Isakov, O., RozenGagnon, K., Arias-Goeta, C., Blanc, H., Beaucourt, S., Haliloglu, T., Schmitt, C., Bonne, I., Ben-Tal, N., Shomron, N., Failloux, A.B., Buchy, P., Vignuzzi, M., 2014. Emergence and transmission of arbovirus evolutionary intermediates with epidemic potential. Cell Host Microbe 15 (6), 706–716. Stetson, D.B., Medzhitov, R., 2006. Type I interferons in host defense. Immunity 25 (3), 373–381. Strauss, J.H., Strauss, E.G., 1994. The alphaviruses: gene expression, replication, and evolution. Microbiol. Rev. 58 (3), 491–562. Takada, A., Kawaoka, Y., 2003. Antibody-dependent enhancement of viral infection: molecular mechanisms and in vivo implications. Rev. Med. Virol. 13 (6), 387– 398. Taylor, A., Sheng, K.C., Herrero, L.J., Chen, W., Rulli, N.E., Mahalingam, S., 2013. Methotrexate treatment causes early onset of disease in a mouse model of Ross River virus-induced inflammatory disease through increased monocyte production. PLoS One 8 (8), e71146. Taylor, K., Kolokoltsova, O., Patterson, M., Poussard, A., Smith, J., Estes, D.M., Paessler, S., 2012. Natural killer cell mediated pathogenesis determines outcome of central nervous system infection with Venezuelan equine encephalitis virus in C3H/HEN mice. Vaccine 30 (27), 4095–4105. Teng, T.S., Foo, S.S., Simamarta, D., Lum, F.M., Teo, T.H., Lulla, A., Yeo, N.K.W., Koh, E.G.L., Chow, A., Leo, Y.S., Merits, A., Chin, K.C., Ng, L.F.P., 2012. Viperin restricts chikungunya virus replication and pathology. J. Clin. Invest. 122 (12), 4447– 4460. Teng, T.S., Kam, Y.W., Tan, J.J., Ng, L.F.P., 2011. Host response to chikungunya virus and perspectives for immune-based therapies. Fut. Virol. 6 (8), 975–984. Teng, T.S., Kam, Y.W., Lee, B., Hapuarachchi, H.C., Wimal, A., Ng, L.C., Ng, L.F.P., 2015. A systematic meta-analysis of immune signatures in patients with acute chikungunya virus infection. J. Infect. Dis. http://dx.doi.org/10.1093/infdis/ jiv049 [epub ahead of print]. Teo, T.H., Her, Z., Tan, J.J.L., Lum, F.M., Lee, W.W.L., Chan, Y.H., Ong, R.Y., Kam, Y.W., Leparc-Goffart, I., Gallian, P., Rénia, L., Delamballerie, X., Ng, L.F.P., 2015. Caribbean and la réunion chikungunya virus isolates differ in their capacity to induce pro-inflammatory Th1 and NK cell responses and acute joint pathology. J. Virol. http://dx.doi.org/10.1128/JVI.00909-15 [epub ahead of print]. Teo, T.H., Lum, F.M., Claser, C., Lulla, V., Lulla, A., Merits, A., Rénia, L., Ng, L.F.P., 2013. A pathogenic role for CD4+ T cells during chikungunya virus infection in mice. J. Immunol. 190 (1), 259–269. Uchime, O., Fields, W., Kielian, M., 2013. The role of e3 in ph protection during alphavirus assembly and exit. J. Virol. 87 (18), 10255–10262. Voss, J.E., Vaney, M.-C., Duquerroy, S., Vonrhein, C., Girard-Blanc, C., Crublet, E., Thompson, A., Bricogne, G., Rey, F.A., 2010. Glycoprotein organization of chikungunya virus particles revealed by X-ray crystallography. Nature 468 (7324), 709–712. Warter, L., Lee, C.Y., Thiagarajan, R., Grandadam, M., Lebecque, S., Lin, R.T., BertinMaghit, S., Ng, L.F., Abastado, J.P., Despres, P., Wang, C.I., Nardin, A., 2011. Chikungunya virus envelope-specific human monoclonal antibodies with broad neutralization potency. J. Immunol. 186 (5), 3258–3264. Watanaveeradej, V., Endy, T.P., Simasathien, S., Kerdpanich, A., Polprasert, N., Aree, C., Vaughn, D.W., Nisalak, A., 2006. Transplacental chikungunya virus antibody kinetics, Thailand. Emerg. Infect. Dis. 12 (11), 1770–1772. Wauquier, N., Becquart, P., Nkoghe, D., Padilla, C., Ndjoyi-Mbiguino, A., Leroy, E.M., 2011. The acute phase of chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J. Infect. Dis. 204 (1), 115–123. Werneke, S.W., Schilte, C., Rohatgi, A., Monte, K.J., Michault, A., Arenzana-Seisdedos, F., Vanlandingham, D.L., Higgs, S., Fontanet, A., Albert, M.L., Lenschow, D.J., 2011. ISG15 is critical in the control of chikungunya virus infection independent of UBE1L mediated conjugation. PLoS Pathog. 7 (10), e1002322. White, L.K., Sali, T., Alvarado, D., Gatti, E., Pierre, P., Streblow, D., Defilippis, V.R., 2011. Chikungunya virus induces ips-1-dependent innate immune activation and protein kinase r-independent translational shutoff. J. Virol. 85 (1), 606–620. Wikan, N., Sakoonwatanyoo, P., Ubol, S., Yoksan, S., Smith, D.R., 2012. Chikungunya virus infection of cell lines: analysis of the east, central and South African lineage. PLoS One 7 (1), e31102. Yun, N.E., Peng, B.H., Bertke, A.S., Borisevich, V., Smith, J.K., Smith, J.N., Poussard, A.L., Salazar, M., Judy, B.M., Zacks, M.A., Estes, D.M., Paessler, S., 2009. Cd4+ t cells provide protection against acute lethal encephalitis caused by Venezuelan equine encephalitis virus. Vaccine 27 (30), 4064–4073. Zellweger, R.M., Prestwood, T.R., Shresta, S., 2010. Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe dengue disease. Cell Host Microbe 7 (2), 128–139. Zhang, Y., Shlomchik, W.D., Joe, G., Louboutin, J.-P., Zhu, J., Rivera, A., Giannola, D., Emerson, S.G., 2002. APCS in the liver and spleen recruit activated allogeneic CD8+ T cells to elicit hepatic graft-versus-host disease. J. Immunol. 169 (12), 7111–7118.
1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076
Please cite this article in press as: Lum, F.-M., Ng, L.F.P. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res. (2015), http:// dx.doi.org/10.1016/j.antiviral.2015.06.009