key: cord-0912070-dp55tcyy authors: Wong, Kai Zhi; Chu, Justin Jang Hann title: The Interplay of Viral and Host Factors in Chikungunya Virus Infection: Targets for Antiviral Strategies date: 2018-05-30 journal: Viruses DOI: 10.3390/v10060294 sha: 756448c2d7dda1e89c0ea4acf6ccd5e2ee02517e doc_id: 912070 cord_uid: dp55tcyy Chikungunya virus (CHIKV) has re-emerged as one of the many medically important arboviruses that have spread rampantly across the world in the past decade. Infected patients come down with acute fever and rashes, and a portion of them suffer from both acute and chronic arthralgia. Currently, there are no targeted therapeutics against this debilitating virus. One approach to develop potential therapeutics is by understanding the viral-host interactions. However, to date, there has been limited research undertaken in this area. In this review, we attempt to briefly describe and update the functions of the different CHIKV proteins and their respective interacting host partners. In addition, we also survey the literature for other reported host factors and pathways involved during CHIKV infection. There is a pressing need for an in-depth understanding of the interaction between the host environment and CHIKV in order to generate potential therapeutics. In recent years, chikungunya virus (CHIKV) has re-emerged as one of the many arthropod-borne viruses (arboviruses) that can pose serious international public health threats [1, 2] . CHIKV is an Alphavirus that belongs to the Togaviridae family and is transmitted mainly by two species of mosquitoes, namely, Aedes albopictus and Aedes aegypti [3] . Chikungunya comes from a Makonde word that refers to the bent-up posture that the disease induces [4] . CHIKV can be classified into three different lineages with distinct genotypes corresponding to their respective geographical origins. They include the Asian, East-Central-South African, and West African genotypes [5] [6] [7] [8] . One unique feature, which distinguishes CHIKV from its arguably well-conserved alphavirus cousins, is its high serum viral loads, which can exceed 10 9 virus particles/mL [9] . This remarkable feature allows easy transmission of the CHIKV to any feeding mosquitos. According to historical records, CHIKV is likely to have been present since 1779 [10] . However, due to the similar clinical manifestations between CHIKV and dengue infections, CHIKV-infected patients were likely to have been initially misdiagnosed as having dengue infection [10, 11] . CHIKV was first isolated and identified during an outbreak in Tanzania in 1953 [4] . Thereafter, many epidemics and outbreaks were documented in a number of African countries in 1958 with thousands of people being infected [12] . Soon after, cases of CHIKV infection mushroomed in many Southeast Asian countries from the 1960s [13] . Between 2005 and 2006, a major unprecedented epidemic of CHIKV swept across many countries within the Indian Ocean territories, where CHIKV was not endemic [13] . motif (UUUUUUA) found on the 6k gene, resulting in a −1 ribosomal frameshifting event [87, 88] . These structural polyproteins are then directed by the signal peptide found on the N-terminus of the E3 protein to the endoplasmic reticulum membrane, where they undergo complete cleavage into individual proteins (pE2 (E3-E2), 6K or TF, and E1) by host proteases [89] . E1 and pE2 proteins associate noncovalently, forming a heterodimer complex, which undergoes posttranslational modifications as it gets shuttled via the Golgi secretory pathway. The host furin cleaves pE2, resulting in a mature E2 viral protein [90] [91] [92] [93] [94] . Soon after, fully developed nucleocapsid cores carrying full-length, positive sense genomes are recruited to the cell plasma membrane where they bud out of the host cells, simultaneously acquiring a portion of the host plasma membrane studded with mature envelope glycoproteins, making up the envelope of the mature virion [53] . It is also interesting to note that in the late phase of infection, another type of cytopathic vacuole called CPV-II can be observed in infected cells [95] [96] [97] . In contrast to CPV-I, CPV-II originate from the trans-Glogi network [98, 99] . Within these vacuoles, the viral envelope glycoproteins (E1 and E2) have been found to be arranged in a hexagonal lattice and packed in arrays of helical tube-like structures [98, 100] . In addition, nucleocapids have also been observed along the periphery of the cytoplasmic side of CPV-II [96, 98, 100] . Given that these structures are in close proximity to the plasma membrane, it is postulated that they aid in viral assembly and/or transport of the envelope proteins to the plasma membrane and viral release via a second mechanism, exocytosis [98, 101] . Structural proteins are known to be involved in processes like entry, fusion, uncoating of virus particle, assembly of virions, and budding. Here, we present a brief review of the functions of each individual viral proteins and their reported interacting host factors. The CHIKV capsid protein is a compact multifunctional protein of 261 amino acids with a molecular weight of about 30 kDa [58, 102, 103] . Unlike the New World encephalitic alphaviruses, the capsid proteins of CHIKV (an Old World arthritogenic alphavirus) do not seem to be involved in the shutting down of the host transcriptional processes [104] . Instead, the Old World arthritogenic viruses rely on nsP2 for inducing host transcriptional and translational shutoff [104] . The capsid protein is made up of three main regions (regions 1, 2, and 3) [105, 106] . Region 1 (1-80 aa) being highly basic in nature (Arg-, Lys-, and Pro-rich), is proposed to be able to bind RNA in a non-specific manner and may also be involved in protein interactions that inhibit host transcription [106] . In contrast, region 2 (81-113 aa) binds specifically to the full length viral RNA genome and also plays an important role in the oligomerisation of other capsid proteins in order to form mature nucleocapsid particles [85, [106] [107] [108] [109] . Region 3 is a serine protease containing a conserved catalytic triad (His 139, Asp 161, and Ser 213) that is able to cleave itself in cis and inactivate itself by binding its active site with its C-terminal tryptophan residue [53, 82, 83, 110] . A recent study reported that the CHIKV capsid is also able to exhibit trans-cleavage properties [111] . In addition, the hydrophobic pocket (containing interacting residues: Val 130, Gly 131, Val 134, Met 135, Trp 245, and Val 250) located on region 3 was found to interact with Pro 404 of the E2 cytoplasmic domain, which is believed to occur during mature particle assembly [102, 106, 112] . Moreover, dimerization of two capsid protein monomers relies on the interaction of the Tyr 186 residue from one monomer with two Asn residues at positions 188 and 220 from the other monomer, all of which are located on region 3 [102] . The CHIKV capsid protein has been reported to possess both one nuclear import (NLS) and two nuclear export signals (NES) (44-53 aa & 143-155 aa) , which allows the protein to traverse actively between the nucleus and cytoplasm [113, 114] . In addition, two host proteins, karyopherin α (Karα) and chromosomal maintenance 1 (CRM1), have been found to be involved in the active nuclear import and export of the CHIKV capsid protein, respectively [113] . Interestingly, both NES are required to be intact for CHIKV capsid to be exported. Mutation of the NES near the N-terminus (44-53 aa) , by replacing the Lys 51 and Met 53 with alanines, resulted in the retention of the CHIKV capsid within the nucleus [114] . Unexpectedly, this lead to the blockage of the host nuclear import system through a mechanism which remains unknown [114] . In addition, Taylor and colleagues showed that mutating the nucleolar localization sequence (NoLs) within the N-terminus results in an attenuated phenotype with smaller plaques and reduced virulence in mice while still being able to elicit an immune response [113] [114] [115] . The precise location of the NLS of the CHIKV capsid has yet to be confirmed. Jacobs and colleagues suggested that the location falls between 1 and 83 aa of the capsid protein, whereas Thomas and colleagues reported that it should be between 60 and 99 aa. Given that the capsid protein is such a crucial, multifunctional viral protein with such an array of functions, more efforts could be channeled to uncover the possible interactions with other potential host factors. E3 proteins carry a signal peptide (a series of polar residues) at their N-terminus, which is crucial for targeting the structural polyprotein towards the endoplasmic reticulum for initial processing [53, 106] . Despite being only 64 amino acids long (~7.4 kDa), it is necessary for the stabilization and maturation of the E2 glycoprotein [58, 87, 116, 117] . From crystal structures, the E3 protein was observed to bind exclusively to the E2 protein [118] . It requires the host furin enzyme to mediate the maturation of the E2 protein by cleaving it in the trans-Golgi system only after dimerization with other available E1 glycoproteins is complete [106, 118] . After cleavage, the E3 protein continues to associate non-covalently with the E1-E2 spikes by relying on the interaction between the Tyr 47 residue on E3 and the Tyr 48 on the E2 [119] . It is not incorporated into the virus and will dissociate when the entire complex is exposed to neutral pH at the plasma membrane surface [119] . Upon dissociation, the acid-sensitive region between the E2 and E1 glycoproteins gets exposed, priming the E1 protein for activation upon contact with low pH during entry [106, 119] . E3 therefore plays an important role in protecting the envelope glycoproteins from low pH and preventing their premature activation. No other host factors that interact with the E3 protein have been reported. The E2 protein (423 aa,~40 kDa), a type I transmembrane glycoprotein, has long been known to be the main antigenic and receptor binding protein for CHIKV [58, 120] . The CHIKV exhibits a wide tropism by being able to replicate in many invertebrate and vertebrate cells [27, 65, [121] [122] [123] . In addition, E2 proteins also serve as stabilizing factors (together with E3 proteins) for the E1-E2 heterodimer during the entire intracellular transport through the secretory pathway, where folding and post-translational modifications take place, and finally to the plasma membrane [106] . To date, no bona fide receptor has been identified for CHIKV. However, there are host factors that have been reported to aid in viral entry. For instance, prohibitin (PHB) was identified to aid in the binding of CHIKV to CHME-5 microglial cells [124] . Wintachai and colleagues also suggested that the possibility of additional co factors that assist the CHIKV entry as poor infection was observed in U937 monocytic cells that also express PHB [124] . In a follow up study, Wintachai and colleagues showed that flavaglines (prohibitin ligand) was able to inhibit CHIKV entry by preventing the CHIKV and prohibitin from colocalising in HEK293T/17 cells [125, 126] . These studies suggest that PHB may be the putative receptor for CHIKV. In addition, ATP synthase β subunit was found to be involved in the entry of CHIKV into Aedes aegypti mosquito cell lines [127] . Additional host factors like protein tyrosine phosphatase non-receptor type 2 (PTPN2), fibril-forming collagen (COL1A2), and actin gamma 1 (ACTG1) were also shown to interact with the E2 proteins in immunoprecipitation experiments [128] . However, further mechanistic studies are required to understand the role of these proteins during CHIKV infection. Jemielity and colleagues showed that overexpression of either human T-cell immunoglobulin and mucin-domain containing proteins 1 (hTIM1) or AXL receptor tyrosine kinase (also known as UFO) of the TAM family of kinases promoted entry of CHIKV by at least 8-fold in HEK293T cells [129] . Their work also showed that CHIKV entry is phosphatidylserine (PS)-dependent, as the PS binding deficient hTIM1 variant did not support viral entry [129] . This suggests that PS is exposed on the membrane of the CHIKV, similar to other enveloped viruses, including Pichinde virus, vesicular stomatitis virus, and vaccinia virus [130, 131] . By taking advantage of the exposed PS, the CHIKV is able to enter the cells. However, the authors noted that the blocking of hTIM1 receptors was less effective in preventing CHIKV entry in Huh7 cells [129] . In addition, there are no reported interactions between hTIM1 and the CHIKV receptor-binding protein, E2. This suggests that the PS-recognising TIM and TAM receptors may not be the bona fide receptors for CHIKV but instead may play a role as attachment factors that enhance CHIKV infectivity. Recent work has shown that only the two surface-exposed domains (domains A and B) of CHIKV E2 are able to bind to cells [132] . Binding of the CHIKV with soluble GAGs was found to be able to inhibit CHIKV infection by up to 90% [132] . In addition, only domain A was able to bind to cell-surface glycosaminoglycans (GAGs)-deficient cells, while domain B was found to interact exclusively with cells expressing GAGs on their cell surface [132] . These results suggest that CHIKV could employ more than one entry mechanism, which most probably explains the wide tropism of cells observed. Therefore, more efforts could be directed to uncover other host factors/potential receptors that could interact with the CHIKV E2 proteins. The TF (~8.3 kDa) protein possess the same N terminus as 6 K (61 amino acids~6.6 kDa) but differs by having a longer basic C terminus (~15 residues) instead of a shorter, hydrophobic C terminus found on 6 K [58, 87, 88] . Unlike other structural proteins, the exact functions of both 6K and TF have not been clearly elucidated [133] . However, studies on other alphaviruses have suggested that these viral accessory proteins mediate membrane permeability and viral budding, and may also be involved in forming ion channels [87, 134, 135] . In addition, both 6 K and TF are also incorporated in low levels into mature virions and are crucial in preserving both the stability and infectivity of the virus [136] [137] [138] . However, no host factor has been found to interact with either of these proteins. The E1 protein (435 amino acids,~44 kDa), a class II viral fusion protein, mediates the fusion of the viral envelope with the host endosomal membrane after endocytosis [58, 120, 139] . This results in the release of the nucleocapsid into the cytoplasm. A single mutation of Ala 226 to a valine residue on the E1 glycoprotein enhanced the dissemination of CHIKV into the secondary organs of Aedes albopictus mosquitos. Moreover, this phenomenon was also detected in the suckling mice [140] . This mutation coincided with the emergence of Aedes albopictus as a second transmission vector during the Indian Ocean epidemic in 2004 [15] . Moreover, in a recent study by Hoornweg and colleagues, this mutation reinforced the cholesterol-dependent membrane fusion of the CHIKV with the host endosomal membranes [66] . In another recent paper, another two epistatic mutations (E1:K211E together with E2:V264A) were also found to notably enhance transmission (62 fold), infection (13 fold), and dissemination (15 fold) in Aedes aegypti mosquitos [141] . However, the exact mechanisms and possible interacting host factors that may facilitate the enhanced fitness of the virus are still unknown. In an attempt to uncover host proteins that interact with E1, Dudha and colleagues performed a yeast two-hybrid (Y2H) screening on a human brain cDNA library [128] . The screen was able to identify 5 interacting host proteins (copper metabolism (Murr1) domain containing 1 (COMMD1), thrombospondin 1 (THBS1), dynein, cytoplasmic 1, heavy chain 1 (DYNC1H1), ATPase Na1/K1 transporting beta 3 polypeptide (ATP1B3), and microtubule-associated protein 1B (MAP1B)). Of these, four hits (COMMD1, THBS1, DYNC1H1, and ATP1B3) passed and were validated via immunoprecipitation and ELISA [128] . However, the biological significance and exact mechanisms of the interactions have yet to be explored. A second study reported that bone marrow stromal antigen 2 (BST-2 or tetherin or CD317) was able to restrict and trap CHIKV on the surface of the host plasma membrane by engaging the E1 glycoprotein [142] . BST-2 knockout mice suggest that BST-2 is able to protect the lymphoid tissues and regulate the inflammatory response induced by the CHIKV. Moreover, another study showed that the longer isoform of BST-2 was found to specifically block the exit of alphaviruses (e.g., SFV and CHIKV) efficiently [143] . In addition, although rubella virus and dengue virus share similar virion structure as the alphaviruses, they responded differently to the presence of BST-2, with the dengue virus not getting inhibited at all [143] . All in all, though much efforts have been focused on uncovering potential interacting host factors, there are still many gaps in our knowledge understanding the interaction of the host factors with the CHIKV structural proteins. The major function of CHIKV non-structural proteins (nsPs) is to replicate the viral genome for translation of structural proteins and for packaging into progeny virions. Aside from this, most nsPs also have additional functions outside of the replication complex. Here, we will present a brief review of the functions of each individual viral protein and their reported interacting non-immunological host factors. The nsP1 (535 amino acids,~60 kDa) protein is responsible for the capping of both the positive-sense genomic viral RNA and the 26S subgenomic RNA [58, 144] . Interestingly, it caps the viral RNA in a non-canonical manner where it first attaches a methyl group (hijacked from the host S-adenosyl-methionine (AdoMet)) to a GTP before transferring the methylated guanylate residue to the nsP2-processed 5 end of the viral RNA [145, 146] . For the capping to be successful, the triphosphates on the 5 end of the viral RNA need to be cleaved by the nsP2 triphosphatase, exposing the diphosphates to allow the transfer to be complete [145] . Capping of the CHIKV RNA is believed to be a strategy to confer protection against degradation by the host exonucleases and also enable efficient translation of the viral mRNA. To date, there is still no high resolution structural information on the nsP1 protein [147] . However, it has been suggested that the capping domain spans across at least the first 400 aa residues from the N terminus [148] . With reference to the secondary structure of the nsP1 of Sindbis virus (SINV), a related alphavirus, the CHIKV nsP1, has been speculated to carry guanylyltransferase activities [149] . However, it is important to note that the sequence (or structural) homology between SINV and CHIKV nsP1 is low, and a crystal structure and further confirmatory biochemical assays would be needed for confirmation of the guanylyltransferase activity of CHIKV nsP1. Another important function of the nsP1 protein is its monotopic interaction with the cytoplasmic side of the plasma membrane bilayer, mediated by its amphipathic alpha helix (approximately between 244 and 263 aa), discovered in a relatively well-studied, close alphavirus relative, the Semliki Forest virus [150, 151] . Additionally, covalent palmitoylation of the nsP1 (417-419 aa) was discovered to be able to strengthen the association with the plasma membrane [149] . This crucial interaction allows the nsP1 to direct and anchor the replication complex to the cell plasma membrane [152] . The interactions between the nsPs have not been well explored. However, there are studies that have shown that nsP1 strongly interacts with nsP4 [153] [154] [155] . Similarly, there is also a lack of studies on the interaction with host proteins. The BST-2 protein was identified to be an anti-viral factor that was downregulated by the nsP1 protein to allow release of the viral particles tethered to the cell surface [142, 143, 156] . The nsP2 protein is one of the more well-studied and also the largest nsP, consisting of 798 amino acids with an approximate molecular weight of 90 kDa [58, 144] . So far, there are no reports of high resolution crystal structures of the entire CHIKV nsP2. However, crystals structures of the CHIKV nsP2 C terminus (~471-791 aa) (PDB code 3TRK (2011) and 4ZTB (2016)) and that of alphavirus relatives, SINV, and Venezuelan equine encephalitis virus (VEEV) are available for comparison [157] [158] [159] . The domains in the N terminal region, however, have been proposed via molecular modelling [160] . Starting from the N terminus, the nsP2 is hypothesized to have three structural domains [160] . The first domain (~1-167 aa) has little homology with other alphaviruses and remains unknown [160] . The subsequent two domains (~168-470 aa) possess characteristic RecA-like domains of superfamily 1 (SF1) group of helicases [160] . The remaining portion (~471-791 aa) holds the protease domain, which can be separated into two smaller sections. The first section bears a papain-like cysteine protease domain (~471-605 aa), while the last section was appears to be a non-functional Ftsj (rrmj) methyltransferase-like domain (~606-791 aa) [160] . This multi-functional nsP2 is capable of performing at least four enzymatic functions. The helicase activity is only functional when the full length nsP2 is available, even though the effector domains are found on the N terminus [160] . Truncated recombinant proteins were found to be inactive [161] . The CHIKV nsP2 is only able to unwind double-stranded RNA that have a 5 overhang of at least 12 bp in and also only in the 5 -3 directionality [160] . Since it possesses RecA-like domains, it is not surprising that it facilitates the complementary base-pairing of single-stranded RNA [160] . The CHIKV nsP2 has also been reported to display nucleotide triphosphatase (NTPase) activities and is able to hydrolyse all dNTPs and NTPs without any preference. However, this is observed only in full length nsP2 [160, 161] . Furthermore, the activation of the NTPase of the nsP2 by RNA and DNA oligonucleotides is, again, only possible with the full length nsP2 [160] . Interestingly, the same active site used for the NTPase activity has also been found to perform RNA 5 triphosphatase activities, the third function, which mediates the cleavage of γ-phosphates from RNA substrates [161] . The last known enzymatic function is the protease activity of the nsP2, which is responsible for processing of the nsP polyprotein [162, 163] . Apart from these enzymatic functions, the nsP2 has been long known to enter the host nucleus during infection, albeit with the absence of any putative nuclear localization signals [147, 164] . Although the exact mechanism is still unknown, the localization of nsP2 is essential in inhibiting the host antiviral responses [165] [166] [167] . One interesting observation reported by Fros and colleagues is that CHIKV nsP2 can be detected in the nuclear region at 4 h post infection (h.p.i) before translocating to the cytoplasm at a later time point of 12 h.p.i [168] . One way that the CHIKV nsP2 is able to induce the shutdown of host transcription is by mediating the degradation of a catalytic subunit of the RNA polymerase II, Rpb1 [169] . The nsP2 protein has also been found to be responsible for the suppression of the host cellular translation processes without affecting viral protein translation [53] . Interactions of the alphavirus nsP2 protein with a large number of ribosomal proteins and proteins that are involved in translations have also been revealed via immunoprecipitation assays [170] . For instance, in VEEV, the ribosomal protein S6 (RpS6) was found to immunoprecipitated with nsP2 in both mammalian and insect cells [171] . Low levels of RpS6 proteins have been correlated strongly with diminished cellular translation activities [171] . However, the exact mechanism of this dephosphorylation phenomenon is still unknown. Recently, nsP2 and nsP3 were also discovered to exhibit RNA interference activity [172] . The promiscuous alphavirus nsP2 is able to bind to not only the other nsPs but also a number of host factors. The interactions with nsPs were confirmed for CHIKV through immunoprecipitation of GST and streptavidin-tagged nsPs and their various domains, and validated via a yeast two-hybird screen and ELISA [173, 174] . In addition, using computational techniques, Rana and colleagues were able to propose a spatial model of the late CHIKV replication complex [173] . Bourai and colleagues performed an extensive study on the host factors interacting with the nsP2 using a yeast two-hybird screen [175] . While they identified 22 unique hits, only heterogeneous nuclear ribonucleoprotein K (hnRNP-K) and ubiquilin 4 (UBQLN4) were observed to play a significant role in CHIKV replication upon gene silencing [175] . Another host factor, the NDP52 human autophagy receptor, was shown to interact with CHIKV nsP2 and acts as a pro-viral factor in human cells [176, 177] . Although the precise mechanism is still unknown, it has been postulated that the human NDP52 interacts with nsP2 in the cytoplasm to prevent the latter from localizing into the nucleus. This could in turn delay cell death, which would allow more time for the CHIKV to replicate [147] . The nsP2 is also implicated in the suppression of the unfolded protein response (UPR) triggered by the production of CHIKV envelope proteins in the endoplasmic reticulum (ER) [168] . Although the exact mechanism of action remains to be clarified, incomplete splicing of the X-box binding protein 1 (XBP1) mRNA (complete splicing is required for UPR) has been observed in cells transfected with nsP2 [168] . Being such a multifunctional protein that is involved in a number of important processes such as viral genome replication and host evasion, CHIKV nsP2 presents an intriguing and promising target for drug development. This has being aptly reviewed by Bakar and Ng [178] . The nsP3 (530 amino acids,~60 kDa) protein possesses three domains, a highly conserved N-terminal macro domain (~160 aa), followed by a zinc 2+ binding domain (~165 aa) and ends with a variable "tail" region (~205 aa) [58, 144, 147] . The macro domain contains a ubiquitous protein module found in all living organisms including positive sense RNA viruses like alphaviruses, coronaviruses, hepatitis E virus and rubella virus [179] . The macro domain is believed to function mainly as an ADP hydrolase that removes mono or poly ADP-ribose marks on proteins. ADP-ribose marks usually occur on Asp, Glu, and Lys residues and are indicative of post-translational modifications by the poly (ADP-ribose) polymerases (PARPs) [180, 181] . In experiments involving mass spectrometry, McPherson and colleagues discovered that the CHIKV nsP3 is able to recognize and hydrolyse the ADP-ribose groups from mono ADP-ribose marks that found on only Asp and Glu residues [182] . The authors went on to show that the loss of the hydrolase significantly compromised the ability of CHIKV to replicate in both baby hamster kidney and Aedes albopictus cells [182] . Moreover, CHIKV infectious clones encoding hydrolases with impaired activities were found to replicate more slowly in mouse neuronal NSC-34 cells with a significant decrease in fitness in neonate mouse model [182] . These results indicate that the macro domain of nsP3 proteins plays an important role in the viral replication and virulence of the CHIKV. In addition, it also suggests that the levels of ADP-ribosylation could play a major role in the anti-viral response by the host. The zinc binding domain of CHIKV nsp3 was inferred from the crystal structure of the SINV by Shin and colleagues [158] . Mutations in this region resulted in the impairment of both the negative-sense and 26S subgenomic RNA synthesis and polyprotein processing in SINV [183, 184] . Similarly, mutations in the zinc-binding region of SFV nsP3 resulted in defects in neurovirulence [185] . However, little is known about their precise mechanisms. The stretch of conserved residues (proline-rich) on the variable "tail" region was found to be able to bind to the host amphiphysin1 and 2 proteins through the Src-homology 3 (SH3) domains found on the amphiphysin proteins [186, 187] . This phenomenon is observed readily upon the transfection of the CHIKV nsP3. Although no validation was performed using CHIKV infected cells, both SFV and SINV infected cells confirmed this observation [186] . Amphiphysins are postulated to be involved in the formation of the spherules (which house replication complexes) given their ability to induce membrane curvature [147, 186, 188] . The nsP3 also binds to GTPase-activating protein (SH3 domain)-binding protein 1 (G3BP1) and its homolog (G3BP2). The G3BPs are found in stress granules, which are believed to have anti-viral properties. For instance, stress granules are involved in the inhibition of RNA translation [147] . By sequestering the G3BPs, the nsP3 is able to prevent stress granules from forming during infection [189] [190] [191] . However, the depletion of the G3BPs was found to be unfavorable for CHIKV replication [192] [193] [194] . The proposed pro-viral activity of the G3BPs includes aiding the switch from the translation of non-structural proteins to viral RNA replication [192] [193] [194] . In mosquito cells, the G3BP homolog, Rasputin (Rin), was found to also exhibit similar phenomenon [195] . However, in cell culture, the silencing of Rin did not adversely affect CHIKV replication [195] . Instead a significant reduction in total infectious viral titer was observed in Aedes albopictus in vivo [195] . Interestingly, Remenyi and colleagues also showed that nsP3 is closely associated with the host cellular lipid droplets and also with nsP1 [196] . Sphingosine kinase 2 (SK2) was reported to co-localize with nsP3 in nsP3 overexpression studies. In addition, SK2 also colocalizes with the CHIKV RNA [197] . Moreover, Reid and colleagues showed that upon SK2 knockdown, there was a significant reduction in infectious viral titer, suggesting a pro-viral role [197] . However, the exact mechanism still remains to be solved. Additionally, there are other host factors (Y-Box-Binding Protein 1 (YBX1), PI3K-Akt-mTOR pathway, DDX1/DDX3, and IKKβ) that were reported to be able to bind or interact with the alphavirus (SINV, SFV, and VEEV) nsP3 reviewed Lark and colleagues [198] . However, these have not been validated for CHIKV. Another pro-viral host protein found to co-immunoprecipitate with the nsP3 is the heat shock protein 90β (Hsp90β), but its exact function is unknown [199] . The bulk of the nsP4 (611 aa,~70 kDa) protein consists of an RNA-dependent RNA polymerase (RdRp) (~500 aa), with the characteristic GDD motif, responsible for viral RNA synthesis at its C-terminus [58, 144, 147] . The remaining~100 aa at its N-terminus, despite being a relatively unknown and seemingly intrinsically disordered stretch of sequences, is still important for the normal function of the nsP4 in SINV [200] . To date, high resolution structures of the CHIKV RdRp are still not available. Recently, Chen and colleagues were able to generate a truncated but soluble, well-folded, functional RdRp catalytic subunit from E. coli [201] . They showed that the CHIKV RdRp has a preference for single-stranded RNA with a 5 -overhang that is of at least 4 nucleotides long [201] . In addition, the RdRp was also found to be rather sensitive to a number of detergents in comparison to the relatively resilient Dengue virus RdRp [201] . The RdRp was also able to exhibit primed extension of templates and terminal adenylyltransferase (TATase) activity regardless of the presence of any template [201] . Similar to the nsP3 protein, the nsP4 protein was found to interact with another Hsp90 protein, Hsp90α. Inhibition of Hsp90α resulted in a decrease in both viral RNA and protein levels [199] . However, the mechanism of the interaction remains to be discovered. In another study, Rathore and colleagues showed that overexpression of the CHIKV nsP4 was able to suppress the phosphorylation of eukaryotic translation initiation factor, alpha subunit (eIF2α), which in turn antagonizes the host unfolded protein response during infection [202] . On the other hand, during SINV infection, rapid phosphorylation of eIF2α was observed instead. However, the mechanism behind this interaction remains unexplored. An in-depth review on the nsP4 functions and interactions was also recently published by Pietila and colleagues [203] . In concert with the above efforts, a number of host factors have been identified via different screening methods. For instance, Paingankar and colleagues discovered that CHIKV interacts with housekeeping proteins like that actin, heat shock protein 70 (HSP70) and STAT2 (Vero-E6 cells only) via virus overlay protein binding assay (VOPBA) complemented with matrix-assisted laser desorption/ionization time of flight analysis (MALDI TOF/TOF) [204] . They went on to show that the silencing of HSP70 resulted in a significant decrease in total infectious viral titer. However, the exact mechanism remains to be solved. Treffers and colleagues employed the use of stable isotope labeling with amino acids in cell culture (SILAC) coupled with liquid chromatography-mass spectrometry (LC-MS) to uncover the temporal dynamics of the cellular response during CHIKV infection [205] . They were able to pick up 13, 38, and 106 proteins that were differentially expressed at 8, 10, and 12 h.p.i, respectively. Moreover, majority of the proteins detected were the subunits of RNA polymerase II, and they were found to be progressively degraded [205] . This is in line with the observation that cellular transcriptional shut off occurs during CHIKV. The authors also reported four anti-viral factors (Rho family GTPase 3 (Rnd3), DEAD box helicase 56 (DDX56), polo-like kinase 1 (Plk1), and ubiquitin-conjugating enzyme E2C (UbcH10)), which were down regulated during CHIKV infection [205] . However, the exact mechanisms remain to be discovered. A very extensive human whole genome siRNA mediated loss-of-function screen was recently performed in a bid to identify effective therapeutics against CHIKV [206] . Karlas and colleagues were able to capture 156 pro-viral and 41 antiviral host factors that affect CHIKV replication [206] . They performed pathway analysis of the identified pro-viral factors and subsequently identified 21 FDA-approved small-molecule inhibitors that were effective against CHIKV by cross referencing with specialized databases [206] . These 21 antiviral compounds were found to act on four host factors (vacuolar-type H+ ATPase (vATPase), CDC-like kinase 1 (CLK1), fms-related tyrosine kinase 4 (FLT4 or VEGFR3), and the K (lysine) acetyltransferase 5 (KAT5 or TIP60)) and two pathways (calmodulin signalling and fatty acid synthesis) [206] . Through in vivo and in vitro work, three of the inhibitors (Tivozanib, Pimozide, and 5-tetradecyloxy-2-furoic acid (TOFA)) were reported to exhibit prophylactic antiviral effects in mice [206] . In addition, when the authors combined two inhibitors (Pimozide and TOFA), each targeting the calmodoulin signalling and fatty acid synthesis pathways, respectively. The synergistic effect resulted in a therapeutic antiviral effect in both in vivo and in vitro studies [206] . However, the exact mechanism and role played by these host pathways in CHIKV replication warrants further work. Nonetheless, the work by Karlas and team showed the importance and relevance of understanding the interplay of host factors during viral infection, as well as the significant translational value that can be gained from performing basic research on the importance of host factors during CHIKV infection. Even though a large number of host factors have been identified through these studies (Table 1) , the mechanistic details of the interplay of the host factors during CHIKV infection are still lacking. The advantages of targeting host factors are plenty, as opposed to targeting just viral proteins. For instance, targeting host factors may allow inhibition of a broad-spectrum of viruses that share same host factors (Table 1) . Moreover, those drugs could also be tested for synergistic effects with specific viral protein inhibitors for development of a more comprehensive treatment plan that targets multiple pathways. This therapeutic approach would also prevent the development of antiviral resistance. For instance, small molecule inhibitors that mimic the interaction sites found on GAGs, PHB, TIM, and TAMs could be developed. These would bind to CHIKV, drastically reduce the opportunity for the CHIKV to interact with the attachment factors present on the host cells and hence dampen its infectivity. Similarly, this approach could also be extended to host factors that are involved in the CHIKV replication cycle like the NDP52 host protein and given in a cocktail to patients. A promising cocktail candidate for clinical trials would be the combination of Pimozide and TOFA concocted by Karlas and team. In drug research, though many drugs screened may have shown to possess great efficacy in vitro, many of them failed during the in vivo validation processes. On the other hand, the cocktail of Pimozide and TOFA was able exhibit impressive efficacy in both in vitro and in vivo studies. Therefore, we feel that there is great potential in this combination host-targeting drug therapy. A group of proteins that transport molecules between the cytoplasm and nucleus. Able to act as either importins or exportins. Binds to NLS of CHIKV capsid protein for nuclear translocation. Proposed to Interact with Middle East Respiratory Syndrome (MERS) virus protein OF4b to prevent NF-kappa-B complex from entering the nucleus. [113, 207] CRM1 (XPO1) Major mammalian export protein that facilitates export of RNA and proteins from the nucleus to the cytoplasm. Pro-viral factor. Binds to the NES/of capsid, allowing exit from the nucleus. Proposed to bind and export the following RNA-containing viral proteins from the nucleus to the cytoplasm: human immunodeficiency virus (HIV) Rev protein cargo complex, human T-cell leukemia virus type 1 (HTLV-1) rex protein, and influenza A ribonucleoprotein complexes. [113, [208] [209] [210] [211] [212] [213] E3 Furin Calcium-dependent serine endoprotease. Preferentially cleaves at sites with paired basic amino acids. Cleaves the E3 protein away from the precursor E2 polyprotein. Shown to be essential for H5N1, H7N1 avian influenza viruses, and Canine distemper virus (CDV). Actual mechanism unknown. Possible attachment/entry factor. Shown to interact with HIV-1 glycoprotein, and the binding is important for its replicative spreading in cells. Interacts with dengue virus E protein and is the first characterized receptor protein for dengue virus in insect cells. Proposed to be entry factors for hepatitis C virus. [124,215-217] A tyrosine phosphatase that dephosphorylates protein tyrosine kinases in both nuclear and cytoplasm compartments. Involved in numerous signaling events (e.g., endocytic recycling). Postulated to be involved in transportation of viral structural proteins to host plasma membrane. Hepatitis C virus nonstructural 3/4A protease cleaves PTPN2 that induces a shift from host Th1 to Th2 immune response. [128, 218] COL1A2 A group 1 collagen found in most connective tissues. Mechanism unknown. Shown to increase infectious viral titer of Sindbis virus (SINV) and also proposed to aid in its transmission. [128,219] Part of cellular trafficking machinery. Postulated to be involved in transportation of viral structural proteins in host cell. The human immunodeficiency virus type 1 (HIV-1) protease was found to cleave actin (ACTG1). Possible attachment/entry factor. Allows binding and infection of hepatitis B virus. Attachment factor for respiratory syncytial virus (RSV), coronavirus NL63 (CoV-NL63), and the severe acute respiratory syndrome coronavirus (SARS-CoV). [132, [221] [222] [223] [224] [225] hTIM1 hTIM1: Involved in regulation of both innate and adaptive immune responses, engulfment of apoptotic cells, and T cell-proliferation. Possible attachment/entry factor. Implicated as receptors for non-enveloped hepatitis A virus and enveloped viruses such as Zaire Ebolavirus and Lake Victoria Marburgvirus. [129, [226] [227] [228] AXL receptor tyrosine kinase Regulate and involved in many important physiological processes like cell proliferation, survival, differentiation, and migration. Possible attachment/entry factor. Implicated as receptors for Ebolavirus, Marburgvirus, pseudo-typed lentiviral, vaccinia virus, and Lassa virus. A proposed scaffold protein that is involved in diverse physiological processes. Able to regulate the ubiquitination and degradation of specific cellular proteins including NF-κB subunit p65. Postulated to be involved in transport of viral structural proteins in host cell and/or involved in regulating host immune response. Enhances latent infection of HIV-1 by stabilizing IκB-α, the inhibitor of NF-κB, and attenuating innate immune response. [128,232,233] Adhesive glycoprotein that binds heparin. Plays a role in dentinogenesis via its anti-angiogenic properties. Also suggested to play a role in ER stress response. Mechanism unknown. Induced by hepatitis C virus (HCV) to promote the proteolytic activation TGF-β1, which promotes HCV RNA replication. [128, 234] Subunit of dynein complex. Integral part of cellular transport machinery across cells including neuronal cells. Plays a role in mitotic spindle and metaphase plate assembly. Postulated to be involved in transport of viral structural proteins in host cell and implicated in neurological manifestations of CHIKV. Aids in uncoating of HIV-1 nucleocapsids during infection. Proposed to be involved in the transport of influenza virus X-31, human foamy virus (HFV), HIV1 reverse transcription complexes (RTC), herpes simplex virus type 1, and Mokola virus. [128, 235, 236] ATP1B3 ATP1B3: Part of the sodium/potassium-transporting ATPase that maintains electrochemical gradient and is important for osmoregulation. Probably facilitates fusion of viral envelope to host membrane during viral entry. Shown to inhibit enterovirus 71 (EV71) replication by up-regulating type-I interferon production. Proposed to be a pro-viral factor for HIV-1 by accelerating the degradation of BST-2. Lipid-raft associated protein that is part of the antiviral response pathway. Blocks the release of many enveloped mammalian virus by tethering the mature virions to the cell plasma membrane of the infected cells. Proposed to restrict virus release by latching onto the CHIKV E1 protein. Restricts Lassa virus replication and release. [ 142, 143, [237] [238] [239] [240] [241] [242] [243] [244] nsP1 BST-2 BST-2: See above. nsP1 reverses BST-2 ability to restrict virus release by down-regulating the latter's expression. See above. [142, 143, 156] nsP2 Rpb1 A catalytic subunit of the RNA polymerase II complex that catalyses RNA transcription. Does not get degraded by the CHIKV nsP2 proteins. Instead is degraded via nsP2 mediated ubiquitination. Same observations were found in Sindbis, Semliki Forest virus (old world alphaviruses). [169] SFRS3/SRp20 (Serine and Arginine Rich Splicing Factor 3) RNA splicing factor, aids in exon-inclusion during alternative splicing. Involved in mRNA nuclear export. Mechanism unknown. Proposed to be crucial for IRES-mediated translation in poliovirus. [175, 245] VIM (Vimentin), TACC3 (transforming, acidic coiled-coil containing protein 3), CEP55 (centrosomal protein 55 kDa), and KLC4 (kinesin light chain 4) VIM, TACC3, CEP55, and KLC4: Cytoskeletal components. Proposed to be hijacked by nsP2 for transport into the infected cells. Interaction with CHIKV nsP3 was also reported and is proposed to aid in the anchorage of the replication complex. Proposed to be involved in the distribution and acidification of endosomes, allowing successful release of influenza A viral genome. TACC3, CEP55, KLC4: Not reported. Part of a complex that regulates and localizes phosphatidylinositol 4-kinase. (PI4K) to the cell plasma membrane. Aids nsP2 in shutting off host cellular processes. Dengue 2 virus induce expressions of proteins that contain tetratricopeptide repeats (TTC). Involved in autophagy; recruits and degrades intracellular pathogens and is able to inhibit proliferations of pathogens like Salmonella. Seems to be able to recruit CHIKV nsP2 and LC3C to the trans-Golgi network that contains double-stranded RNA and other nsPs. This is postulated to allow formation of the replication complexes, thereby promoting viral infection. Proposed to modulate innate immune response upon interacting with influenza virus protein PB1-F2. Regulates cell cycle and is directly involved in cellular proliferation, quiescence, and cancer. Drives the internalization of the replication complex. Essential for survival of host and virus (Hepatitis C Virus, Vaccinia, and Cowpox Virus). [75, 263, 264] G3BP1 & G3BP2 Marker for stress granules and may be an effector for stress granule assembly. Able to unwind DNA and RNA. Postulated to be a scaffold protein which could transport mRNA. Colocalizes with nsP2 and nsP3. Depletion of both proteins results in reduction of viral RNA (especially the negative sense RNA), proteins, and infectious titer. Authors proposed that the G3BPs could mediate the switch from translation to amplification of viral genome. Knock-down of G3BP1 results an increase in HIV-1 viral titer only in primary T cells and macrophages. Found to interact with HIV-1 RNA in the cytoplasm. G3BP1 is proposed to sequester viral RNA transcripts, preventing translation and packaging. G3BP1 and G3BP2: DENV-2 non-coding subgenomic flaviviral RNA (sfRNA) was found to bind to both G3BP1 and G3BP2 and inhibit their antiviral activities. [192, 265, 266] SK2 A lipid kinase that phosphorylates sphingosine to form sphingosine 1-phosphate (SPP). Involved in cell differentiation, growth, and host immunity. Colocalizes with CHIKV RNA and nsPs. Knock-down of SK2 inhibits CHIKV infection. Not reported. [197] Hsp90β A cytoplasmic isoform of HSP-90s molecular chaperons that is constitutively expressed. They are able to modulate different cellular processes (e.g., refold proteins) to maintain cellular homeostasis. Mechanism unclear. Proposed to have an ancillary role in CHIKV replication. On a side note, Hsp90 is proposed to stabilize CHIKV nsP2 during infection. Proposed to be the binding receptor for Japanese encephalitis virus (JEV). Proposed to facilitate assembly of enterovirus 71 viral particles. Hepatitis B virus polymerase interacts with Hsp90β to suppress NF-kB signaling. [199, [267] [268] [269] [270] nsP4 LCP1 (Lymphocyte cytosolic protein 1/L-Plastin) Binds to actin and aids in activation of T-cells during co-stimulation through other receptors. Mechanism unknown. Not reported. [175] Hsp90α: A cytoplasmic isoform of HSP-90s molecular chaperons that is produced during cell stress response. They are able to modulate different cellular processes (e.g., refold proteins) to maintain cellular homeostasis. Proposed to help in stabilizing CHIKV nsP4 and aid in formation of CHIKV replication complex in the cytosol. Knock-down of Hsp90α resulted in a decrease in viral RNA. Hsp90α: Not reported. [199] eIF2α eIF2α: A eukaryotic initiation factor that is essential for initiating translation. nsP4 suppresses the serine-51 phosphorylation of eIF2α, which in turn regulates the PERK pathway, allowing the CHIKV to overcome the host unfolded protein response machinery. The presence of the three rotavirus proteins, VP2, NSP2, and NSP5, induces the phosphorylation of eIF2α. However, formation of stress granules (which stalls translation) was inhibited. HSV utilizes its neurovirulence factor ICP34.5 to dephosphorylate eIF2α. It should still be noted that targeting host factors comes with the risk of toxicity, especially when these host factors perform vital functions in the host cells. Design of therapeutics therefore needs to be optimized to target interactions between host and viral factors, while minimizing disruption of essential cellular processes. With the significant bottlenecks in our knowledge of basic CHIKV virology and its interacting host partners, more research is needed to understand the molecular mechanisms of the interactions between the CHIKV and its host factors. This would not only help to increase the knowledge pool but also provide more opportunities and avenues to develop, optimize, and/or speed up the production or repurposing of potential therapeutics to combat this medically important re-emerging arbovirus. Author Contributions: K.Z.W. and J.J.H.C. wrote the paper. Chikungunya virus: Epidemiology, replication, disease mechanisms, and prospective intervention strategies How Great is the Threat of Chikungunya Virus? Genetic characterization of 2006-2008 isolates of Chikungunya virus from Kerala, South India, by whole genome sequence analysis An epidemic of virus disease in Southern Province, Tanganyika Territory Re-emergence of chikungunya and o'nyong-nyong viruses: Evidence for distinct geographical lineages and distant evolutionary relationships Changing patterns of chikunya virus: Re-emergence of a zoonotic arbovirus Genome microevolution of chikungunya viruses causing the Indian Ocean outbreak Genome-Scale Phylogenetic Analyses of Chikungunya Virus Reveal Independent Emergences of Recent Epidemics and Various Evolutionary Rates Chikungunya fever: CNS infection and pathologies of a re-emerging arbovirus Chikungunya and dengue: A case of mistaken identity? Chikungunya virus infections among patients with dengue-like illness at a tertiary care hospital in the Philippines Chikungunya: An overview Chikungunya fever: An epidemiological review of a re-emerging infectious disease Chikungunya virus infection during pregnancy Two Chikungunya isolates from the outbreak of La Reunion (Indian Ocean) exhibit different patterns of infection in the mosquito, Aedes albopictus Global expansion of chikungunya virus: Mapping the 64-year history Chikungunya: Bending over the Americas and the rest of the world Infection with chikungunya virus in Italy: An outbreak in a temperate region Diseases Surveillance Epidemiology of Chikungunya Virus Outbreaks in Guadeloupe and Martinique, 2014: An Observational Study in Volunteer Blood Donors Outbreak of Chikungunya in the French Caribbean Islands of Martinique and Guadeloupe: Findings from a Hospital-Based Surveillance System European Centre for Disease Prevention and Control. Clusters of Autochthonous Chikungunya Cases in Conclusions and Options for Response Autochthonous Chikungunya transmission and extreme climate events in Southern France Rheumatoid arthritic syndrome after chikungunya fever A mouse model for Chikungunya: Young age and inefficient type-I interferon signaling are risk factors for severe disease Human Muscle Satellite Cells as Targets of Chikungunya Virus Infection Focus on Chikungunya pathophysiology in human and animal models. Microbes Infect Chikungunya Fever: A Clinical and Virological Investigation of Outpatients on Reunion Island Chikungunya disease: Infection-associated markers from the acute to the chronic phase of arbovirus-induced arthralgia Congenital and perinatal complications of chikungunya fever: A Latin American experience Transmission materno-foetale précoce du virus Chikungunya Travail original Infection à Chikungunya chez la femme enceinte et risque de transmission materno-foetale Chikungunya virus: An update on the biology and pathogenesis of this emerging pathogen Clinical progression of chikungunya fever during acute and chronic arthritic stages and the changes in joint morphology as revealed by imaging Post-epidemic Chikungunya disease on reunion island: Course of rheumatic manifestations and associated factors over a 15-month period Amelioration of alphavirus-induced arthritis and myositis in a mouse model by treatment with bindarit, an inhibitor of monocyte chemotactic proteins Chikungunya disease in nonhuman primates leads to long-term viral persistence in macrophages Chikungunya virus: An update on antiviral development and challenges Antiviral Treatment of Chikungunya Virus Infection Chloroquine phosphate treatment of chronic Chikungunya arthritis. An open pilot study Ribavirin therapy for Chikungunya arthritis UK-427,857), a Potent, Orally Bioavailable, and Selective Small-Molecule Inhibitor of Chemokine Receptor CCR5 with Broad-Spectrum Anti-Human Immunodeficiency Virus Type 1 Activity Cyclosporin A inhibits the replication of diverse coronaviruses New treatments for influenza The development of new therapies for human herpesvirus 6 Progress in the development of new therapies for herpesvirus infections Combatting Emerging Viral Threats Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection Curing a viral infection by targeting the host: The example of cyclophilin inhibitors Productive Replication of Ebola Virus Is Regulated by the c-Abl1 Tyrosine Kinase The alphaviruses: Gene expression, replication, and evolution Structural proteins of Chikungunya virus Electron microscope study of development of Chikungunya virus in green monkey kidney stable (VERO) cells Replication cycle of chikungunya: A re-emerging arbovirus Replication of alphaviruses: A review on the entry process of alphaviruses into cells Complete nucleotide sequence of chikungunya virus and evidence for an internal polyadenylation site Chapter 29 Chikungunya Virus Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography Biology and pathogenesis of chikungunya virus Characterization of reemerging chikungunya virus Endocytosis of chikungunya virus into mammalian cells: Role of clathrin and early endosomal compartments Genome-Wide RNAi Screen Identifies Novel Host Proteins Required for Alphavirus Entry Early Events in Chikungunya Virus Infection-From Virus CellBinding to Membrane Fusion Dynamics of Chikungunya Virus Cell Entry Unraveled by Single-Virus Tracking in Living Cells Mosquito Cellular Factors and Functions in Mediating the Infectious entry of Chikungunya Virus Chikungunya virus fusion properties elucidated by single-particle and bulk approaches Role of ribosomes in Semliki Forest virus nucleocapsid uncoating The signal for translational readthrough of a UGA codon in Sindbis virus RNA involves a single cytidine residue immediately downstream of the termination codon Versatile trans-replication systems for chikungunya virus allow functional analysis and tagging of every replicase protein Functional Sindbis Virus Replicative Complexes Are Formed at the Plasma Membrane Biogenesis of the Semliki Forest Virus RNA Replication Complex Phosphatidylinositol 3-Kinase-, Actin-, and Microtubule-Dependent Transport of Semliki Forest Virus Replication Complexes from the Plasma Membrane to Modified Lysosomes Differential Phosphatidylinositol-3-Kinase-Akt-mTOR Activation by Semliki Forest and Chikungunya Viruses Is Dependent on nsP3 and Connected to Replication Complex Internalization Proteolytic processing of semliki forest virus-specif non-structural polyprotein Cleavage-site preferences of Sindbis Polypeptide requirements for assembly of functional Sindbis virus replication complexes: A model for the temporal regulation of minus-and plus-strand RNA synthesis Sindbis virus RNA-negative mutants that fail to convert from minus-strand to plus-strand synthesis: Role of the nsP2 protein Regulation of Sindbis Virus RNA Replication: Uncleaved P123 and nsP4 Function in Minus-Strand RNA Synthesis, whereas Cleaved Products from P123 Are Required for Efficient Plus-Strand RNA Synthesis Regulation of Semliki Forest virus RNA replication: A model for the control of alphavirus pathogenesis in invertebrate hosts Processing of the Semliki Forest virus structural polyprotein: Role of the capsid protease Evidence for an autoprotease activity of sindbis virus capsid protein Assembly of the Semliki Forest Virus Membrane Glycoproteins in the Membrane of the Endoplasmatic Reticulum in Vitro Identification of a region in the Sindbis virus nucleocapsid protein that is involved in specificity of RNA encapsidation Evidence for specificity in the encapsidation of Sindbis virus RNAs Discovery of frameshifting in Alphavirus 6K resolves a 20-year enigma Functional Characterization of the Alphavirus TF Function of Semliki Forest virus E3 peptide in virus assembly: Replacement of E3 with an artificial signal peptide abolishes spike heterodimerization and surface expression of E1 Effects of site-directed mutations of transmembrane cysteines in sindbis virus E1 and E2 glycoproteins on palmitylation and virus replication Site-directed mutations in the Sindbis virus E2 glycoprotein identify palmitoylation sites and affect virus budding Formation of the Semlike Forest membrane glycoprotein complexes in the infected cell Dissection of Semliki Forest virus glycoprotein delivery from the trans-Golgi network to the cell surface in permeabilized BHK cells Communication of post-Golgi elements with early endocytic pathway: Regulation of endoproteolytic cleavage of Semliki Forest virus p62 precursor Membrane protein lateral interactions control Semliki Forest virus budding The dynamic nature of the Golgi complex Dissection of the golgi-complex.1. Monensin inhibits the transport of viral membrane-proteins from medial to trans golgi cisternae in baby hamster-kidney cells infected with Semliki Forest virus Structural Evidence of Glycoprotein Assembly in Cellular Membrane Compartments prior to Alphavirus Budding Comparative analysis of the genome sequences and replication profiles of chikungunya virus isolates within the East, Central and South African (ECSA) lineage Interactions between the transmembrane segments of the alphavirus E1 and E2 proteins play a role in virus budding and fusion Transport and budding at two distinct sites of visible nucleocapsids of West Nile (Sarafend) virus Inhibition of chikungunya virus by picolinate that targets viral capsid protein Functional dissection of the alphavirus capsid protease: Sequence requirements for activity The Old World and New World Alphaviruses Use Different Virus-Specific Proteins for Induction of Transcriptional Shutoff Alphavirus Capsid Protein Helix I Controls a Checkpoint in Nucleocapsid Core Assembly Function of Chikungunya Virus Structural Proteins Interactions between sindbis virus RNAs and a 68 amino acid derivative of the viral capsid protein further defines the capsid binding site Sindbis virus nucleocapsid assembly: RNA folding promotes capsid protein dimerization Identification of Interactions between Sindbis Virus Capsid Protein and Cytoplasmic vRNA as Novel Virulence Determinants Structure of Sindbis virus core protein reveals a chymotrypsin-like serine proteinase and the organization of the virion Kinetic characterization of trans-proteolytic activity of Chikungunya virus capsid protease and development of a FRET-based HTS assay Structure-function insights into chikungunya virus capsid protein: Small molecules targeting capsid hydrophobic pocket Chikungunya virus capsid protein contains nuclear import and export signals Mutation of a conserved nuclear export sequence in chikungunya virus capsid protein disrupts host cell nuclear import Mutation of the N-Terminal Region of Chikungunya Virus Capsid Protein: Implications for Vaccine Design The Alphavirus E3 Glycoprotein Functions in a Clade-Specific Manner Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells Structural changes of envelope proteins during alphavirus fusion The Role of E3 in pH Protection during Alphavirus Assembly and Exit Expression and evaluation of Chikungunya virus E1 and E2 envelope proteins for serodiagnosis of chikungunya virus infection Characterization of Chikungunya pseudotyped viruses: Identification of refractory cell lines and demonstration of cellular tropism differences mediated by mutations in E1 glycoprotein Establishment of a Novel Primary Human Skeletal Myoblast Cellular Model for Chikungunya Virus Infection and Pathogenesis PH-dependent entry of chikungunya virus into Aedes albopictus cells Identification of Prohibitin as a Chikungunya Virus Receptor Protein Assessment of flavaglines as potential chikungunya virus entry inhibitors Prohibitin ligands in cell death and survival: Mode of action and therapeutic potential Involvement of ATP synthase β subunit in chikungunya virus entry into insect cells Host-pathogen interactome analysis of Chikungunya virus envelope proteins E1 and E2 TIM-family Proteins Promote Infection of Multiple Enveloped Viruses through Virion-associated Phosphatidylserine Vaccinia virus uses macropinocytosis and apoptotic mimicry to enter host cells Targeting Inside-Out Phosphatidylserine as a Therapeutic Strategy For Viral Diseases Identification of Functional Determinants in the Chikungunya Virus E2 Protein Disentangling the frames, the state of research on the alphavirus 6K and TF proteins Interfacial domains in sindbis virus 6K protein: Detection and functional characterization The 6-kilodalton membrane protein of Semliki Forest virus is involved in the budding process The sindbis virus 6K protein can be detected in virions and is acylated with fatty acids Fate of the 6K membrane protein of semliki forest virus during virus assembly Effects of an In-Frame Deletion of the 6k Gene Locus from the Genome of Ross River Virus Cell-based analysis of Chikungunya virus E1 protein in membrane fusion A single mutation in Chikungunya virus affects vector specificity and epidemic potential Two novel epistatic mutations (E1:K211E and E2:V264A) in structural proteins of Chikungunya virus enhance fitness in Aedes aegypti BST-2/tetherin-mediated restriction of chikungunya (CHIKV) VLP budding is counteracted by CHIKV non-structural protein 1 (nsP1) BST2/Tetherin inhibition of alphavirus exit Alphavirus RNA synthesis and non-structural protein functions Reaction in alphavirus mRNA capping: Formation of a covalent complex of nonstructural protein nsPl with 7-methyl-GMP Conventional and unconventional mechanisms for capping viral mRNA Functions of Chikungunya Virus Nonstructural Proteins Sequence analysis reveals a conserved extension in the capping enzyme of the alphavirus supergroup, and a homologous domain in nodaviruses The effects of palmitoylation on membrane association of Semliki Forest virus RNA capping enzyme Membrane binding mechanism of an RNA virus-capping enzyme Semliki Forest virus mRNA capping enzyme requires association with anionic membrane phospholipids for activity Role of the Amphipathic Peptide of Semliki Forest Virus Replicase Protein nsP1 in Membrane Association and Virus Replication Modification of Asn374 of nsP1 suppresses a Sindbis virus nsP4 minus-strand polymerase mutant Suppressor mutations that allow Sindbis virus RNA polymerase to function with nonaromatic amino acids at the N-terminus: Evidence for interaction between nsP1 and nsP4 in minus-strand RNA synthesis Mutations at the palmitoylation site of non-structural protein nsP1 of Semliki Forest virus attenuate virus replication and cause accumulation of compensatory mutations Critical role for bone marrow stromal antigen 2 in acute Chikungunya virus infection The Crystal Structure of the Venezuelan Equine Encephalitis Alphavirus nsP2 Protease Structural and functional insights into alphavirus polyprotein processing and pathogenesis Identification of chikungunya virus nsP2 protease inhibitors using structure-base approaches Functional cross-talk between distant domains of chikungunya virus non-structural protein 2 is decisive for its RNA-modulating activity NTPase and 5 -RNA triphosphatase activities of chikungunya virus nsP2 protein Structure-function relationship of Chikungunya nsP2 protease: A comparative study with papain Site-specific Protease Activity of the Carboxyl-terminal Domain of Semliki Forest Virus Replicase Protein nsP2 Nuclear localization of Semliki Forest virus-specific nonstructural protein nsP2 Chikungunya Virus Nonstructural Protein 2 Inhibits Type I/II Interferon-Stimulated JAK-STAT Signaling Roles of Nonstructural Protein nsP2 and α/β Interferons in Determining the Outcome of Sindbis Virus Infection Roles of Nonstructural Protein nsP2 and α/β Interferons in Determining the Outcome of Sindbis Virus Infection Semliki Forest Virus Nonstructural Protein 2 Is Involved in Suppression of the Type I Interferon Response Chikungunya virus non-structural protein 2-mediated host shut-off disables the unfolded protein response Evasion of the Innate Immune Response: The Old World Alphavirus nsP2 Protein Induces Rapid Degradation of Rpb1, a Catalytic Subunit of RNA Polymerase II Development of Sindbis Viruses Encoding nsP2/GFP Chimeric Proteins and Their Application for Studying nsP2 Functioning Ribosomal Protein S6 Associates with Alphavirus Nonstructural Protein 2 and Mediates Expression from Alphavirus Messages Analysis of chikungunya virus proteins reveals that non-structural proteins nsP2 and nsP3 exhibit RNA interference (RNAi) suppressor activity Network mapping among the functional domains of Chikungunya virus nonstructural proteins Mapping interactions of Chikungunya virus nonstructural proteins Mapping of Chikungunya Virus Interactions with Host Proteins Identified nsP2 as a Highly Connected Viral Component Chikungunya triggers an autophagic process which promotes viral replication Species-specific impact of the autophagy machinery on Chikungunya virus infection Nonstructural Proteins of Alphavirus-Potential Targets for Drug Development The Crystal Structures of Chikungunya and Venezuelan Equine Encephalitis Virus nsP3 Macro Domains Define a Conserved Adenosine Binding Pocket Family-wide analysis of poly(ADP-ribose) polymerase activity The Promise of Proteomics for the Study of ADP-Ribosylation ADP-ribosylhydrolase activity of Chikungunya virus macrodomain is critical for virus replication and virulence Deletion and duplication mutations in the C-terminal nonconserved region of Sindbis virus nsP3: Effects on phosphorylation and on virus replication in vertebrate and invertebrate cells Functional Analysis of nsP3 Phosphoprotein Mutants of Sindbis Virus Functional Analysis of nsP3 Phosphoprotein Mutants of Sindbis Virus Amino acid mutations in the replicase protein nsP3 of Semliki Forest virus cumulatively affect neurovirulence SH3 domain-mediated recruitment of host cell amphiphysins by alphavirus nsp3 promotes viral RNA replication Structural basis of the high affinity interaction between the Alphavirus nonstructural protein-3 (nsP3) and the SH3 domain of amphiphysin-2 Membrane curvature and its generation by BAR proteins Chikungunya Virus nsP3 Blocks Stress Granule Assembly by Recruitment of G3BP into Cytoplasmic Foci The C-Terminal Repeat Domains of nsP3 from the Old World Alphaviruses Bind Directly to G3BP Sequestration of G3BP coupled with efficient translation inhibits stress granules in Semliki Forest virus infection Stress Granule Components G3BP1 and G3BP2 Play a Proviral Role Early in Chikungunya Virus Replication New World and Old World Alphaviruses Have Evolved to Exploit Different Components of Stress Granules, FXR and G3BP Proteins, for Assembly of Viral Replication Complexes Combined structural, biochemical and cellular evidence demonstrates that both FGDF motifs in alphavirus nsP3 are required for efficient replication Mosquito Rasputin interacts with chikungunya virus nsP3 and determines the infection rate in Aedes albopictus SNAP-Tagged Chikungunya Virus Replicons Improve Visualisation of Non-Structural Protein 3 by Fluorescence Microscopy Sphingosine kinase 2 is a chikungunya virus host factor co-localized with the viral replication complex Interactions of Alphavirus nsP3 protein with host proteins Chikungunya virus nsP3 & nsP4 interacts with HSP-90 to promote virus replication: HSP-90 inhibitors reduce CHIKV infection and inflammation in vivo Requirement for the Amino-Terminal Domain of Sindbis Virus nsP4 during Virus Infection Chikungunya virus nsP4 RNA-dependent RNA polymerase core domain displays detergent-sensitive primer extension and terminal adenylyltransferase activities Differential unfolded protein response during Chikungunya and Sindbis virus infection: CHIKV nsP4 suppresses eIF2a phosphorylation Alphavirus polymerase and RNA replication Identification of chikungunya virus interacting proteins in mammalian cells Temporal SILAC-based quantitative proteomics identifies host factors involved in chikungunya virus replication A human genome-wide loss-of-function screen identifies effective chikungunya antiviral drugs Sola, I. MERS-CoV 4b protein interferes with the NF-κB-dependent innate immune response during infection A novel nucleoporin-like protein interacts with both HIV-1 Rev nuclear export signal and CRM-1 Interaction of the Influenza Virus Nucleoprotein with the Cellular CRM1-Mediated Nuclear Export Pathway The carboxy-terminal region of the human immunodeficiency virus type 1 protein Rev has multiple roles in mediating CRM1-related Rev functions A multifunctional domain in human CRM1 (exportin 1) mediates RanBP3 binding and multimerization of human T-cell leukemia virus type 1 Rex protein Requirement of DDX3 DEAD box RNA helicase for HIV-1 Rev-RRE export function Kinetic and Molecular Analysis of Nuclear Export Factor CRM1 Association with Its Cargo In Vivo Kinetic and Molecular Analysis of Nuclear Export Factor CRM1 Association with Its Cargo In Vivo Identification and characterization of prohibitin as a receptor protein mediating DENV-2 entry into insect cells A Novel Class of Small Molecule Compounds that Inhibit Hepatitis C Virus Infection by Targeting the Prohibitin-CRaf Pathway Identification of the Cellular Prohibitin 1/Prohibitin 2 Heterodimer as an Interaction Partner of the C-Terminal Cytoplasmic Domain of the HIV-1 Glycoprotein Cleavage of the T Cell Protein Tyrosine Phosphatase by the Hepatitis C Virus Nonstructural 3/4A Protease Induces a Th1 to Th2 Shift Reversible by Ribavirin Therapy Effects of collagen matrix on Sindbis virus infection of BHK cells HIV-1 protease cleaves actin during acute infection of human T-lymphocytes The Fusion Glycoprotein of Human Respiratory Syncytial Virus Facilitates Virus Attachment and Infectivity via an Interaction with Cellular Heparan Sulfate Neutralization of hepatitis A virus (HAV) by an immunoadhesin containing the cysteine-rich region of HAV cellular receptor-1 Human Coronavirus NL63 Utilizes Heparan Sulfate Proteoglycans for Attachment to Target Cells Role of glycosaminoglycans for binding and infection of hepatitis B virus Glycosaminoglycans and infection T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for Zaire Ebolavirus and Lake Victoria Marburgvirus Identification of a surface glycoprotein on African green monkey kidney cells as a receptor for hepatitis A virus The human homolog of HAVcr-1 codes for a hepatitis A virus cellular receptor Tyro3 Family-Mediated Cell Entry of Ebola and Marburg Viruses Identification of Cell Surface Molecules Involved in Dystroglycan-Independent Lassa Virus Cell Entry The Soluble serum protein gas6 bridges virion envelope phosphatidylserine to the TAM receptor tyrosine kinase Axl to mediate viral entry COMMD proteins, a novel family of structural and functional homologs of MURR1 COMMD1/Murr1 Reinforces HIV-1 Latent Infection through IκB-α Stabilization Hepatitis C virus-induced furin and thrombospondin-1 activate TGF-β1: Role of TGF-β1 in HCV replication A superhighway to virus infection HIV-1 Uncoating Is Facilitated by Dynein and Kinesin 1 Influenza virus is not restricted by tetherin whereas influenza VLP production is restricted by tetherin Modulation of hepatitis C virus release by the interferon-induced protein BST-2/tetherin Tetherin-mediated restriction of filovirus budding is antagonized by the Ebola glycoprotein Infectious Lassa Virus, but Not Filoviruses, Is Restricted by BST-2/Tetherin Interferon-Induced Cell Membrane Proteins, IFITM3 and Tetherin, Inhibit Vesicular Stomatitis Virus Infection via Distinct Mechanisms The RING-CH Ligase K5 Antagonizes Restriction of KSHV and HIV-1 Particle Release by Mediating Ubiquitin-Dependent Endosomal Degradation of Tetherin Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu Tetherin Inhibits HIV-1 Release by Directly Tethering Virions to Cells Viral Proteinase Requirements for the Nucleocytoplasmic Relocalization of Cellular Splicing Factor SRp20 during Picornavirus Infections Proteomic profiling of chikungunya virus-infected human muscle cells: Reveal the role of cytoskeleton network in CHIKV replication Vimentin plays a role in the release of the influenza A viral genome from endosomes Identification of TRIM27 as a Novel Degradation Target of Herpes Simplex Virus 1 ICP0 EWSR1 Binds the Hepatitis C Virus cis-Acting Replication Element and Is Required for Efficient Viral Replication Activates the B-myb Promoter through Formation of a Nuclear Complex with the Retinoblastoma Protein and the Novel Protein PAM14 Identification of HNRNPK as Regulator of Hepatitis C Virus Particle Production Heterogeneous Nuclear Ribonucleoprotein K Supports Vesicular Stomatitis Virus Replication by Regulating Cell Survival and Cellular Gene Expression Distinct expression of interferon-induced protein with tetratricopeptide repeats (IFIT) 1/2/3 and other antiviral genes between subsets of dendritic cells induced by dengue virus 2 infection Mumps virus small hydrophobic protein targets ataxin-1 ubiquitin-like interacting protein (ubiquilin 4) Ubiquitin Ligase WWP1 Interacts with Ebola Virus VP40 to Regulate Egress p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PL pro via E3 ubiquitin ligase RCHY1 Functional analysis of Tpr: Identification of nuclear pore complex association and nuclear localization domains and a role in mRNA export Measles virus-induced disruption of the glial-fibrillary-acidic protein cytoskeleton in an astrocytoma cell line (U-251) Avian reovirus protein p17 functions as a nucleoporin Tpr suppressor leading to activation of p53, p21 and PTEN and inactivation of PI3K/AKT/mTOR and ERK signaling pathways Influenza virus protein PB1-F2 interacts with CALCOCO2 (NDP52) to modulate innate immune response Macroautophagy-Friend or foe of viral replication? Activation of the N-Ras-PI3K-Akt-mTOR Pathway by Hepatitis C Virus: Control of Cell Survival and Viral Replication Activation of the PI3K/Akt Pathway Early during Vaccinia and Cowpox Virus Infections Is Required for both Host Survival and Viral Replication A. G3BP1 restricts HIV-1 replication in macrophages and T-cells by sequestering viral RNA G3BP2 and CAPRIN1 Are Required for Translation of Interferon Stimulated mRNAs and Are Targeted by a Dengue Virus Non-coding RNA Heat shock protein-90-beta facilitates enterovirus 71 viral particles assembly Heat shock protein 90 positively regulates Chikungunya virus replication by stabilizing viral non-structural protein nsP2 during infection Heat shock protein 90β in the Vero cell membrane binds Japanese encephalitis virus Hepatitis B virus polymerase suppresses NF-κB signaling by inhibiting the activity of IKKs via interaction with Hsp90β Rotavirus infection induces the phosphorylation of eIF2α but prevents the formation of stress granules Increased eIF2α phosphorylation attenuates replication of herpes simplex virus 2 vhs mutants in mouse embryonic fibroblasts and correlates with reduced accumulation of the PKR antagonist ICP34.5 We would like to thank Parveen Kaur for her tireless proof-reading. We acknowledge support from MOE Tier 2 2017 (MOE 2017-T2-1-078) awarded to Justin Jang Hann Chu. Due to the space constraints, we apologize for not being able to include all studies that have been reported. The authors declare no conflict of interest.